WO1991017424A1 - Intracellular delivery of biologically active substances by means of self-assembling lipid complexes - Google Patents

Intracellular delivery of biologically active substances by means of self-assembling lipid complexes Download PDF

Info

Publication number
WO1991017424A1
WO1991017424A1 PCT/US1991/002962 US9102962W WO9117424A1 WO 1991017424 A1 WO1991017424 A1 WO 1991017424A1 US 9102962 W US9102962 W US 9102962W WO 9117424 A1 WO9117424 A1 WO 9117424A1
Authority
WO
WIPO (PCT)
Prior art keywords
lipid
negatively charged
bioactive substance
composition
vesicles
Prior art date
Application number
PCT/US1991/002962
Other languages
French (fr)
Inventor
Philip L. Felgner
Original Assignee
Vical, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Vical, Inc. filed Critical Vical, Inc.
Publication of WO1991017424A1 publication Critical patent/WO1991017424A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids

Definitions

  • the present invention relates to methods that are used to enhance delivery of "biologically and pharmacologically active agents, particularly polynucleotides, proteins, peptides, and drug molecules, by facilitating transmembrane transport or by encouraging adhesion to biological surfaces. It relates particularly to self-assembling systems, comprising lipids having cationic charged regions, that facilitate intracellular delivery of these bioactive agents.
  • bioactive agents need to enter cells to exert their biological effect. Some are able to act on cell surfaces through cell surface receptors, while the effect of others is mediated through an interaction with extracellular components.
  • many valuable therapeutic agents that are most effective in influencing cell function at the subcellular or molecular level comprising natural biological molecules and their analogues, or foreign substances such as drugs, are preferably incorporated within the cell in order to produce their effect.
  • the cell membrane provides a selective barrier which is impermeable to many of these agents.
  • the complex composition of the cell membrane comprises phospholipids, glycolipids, and cholesterol, as well as intrinsic and extrinsic proteins, and its functions are further influenced by cytoplas ic components, comprising small ions and subcellular structures.
  • Therapeutic peptides that could be introduced in this way include lymphokines, such as interleukin-2, tumor necrosis factor, the interferons, growth factors, such as nerve growth factor, epidermal growth factor, and human growth hormone, tissue plasminogen activator, factor VIII:C, erythropoietin, insulin, calcitonin, and amylin.
  • lymphokines such as interleukin-2, tumor necrosis factor, the interferons
  • growth factors such as nerve growth factor, epidermal growth factor, and human growth hormone
  • tissue plasminogen activator such as erythropoietin
  • insulin calcitonin
  • amylin amylin.
  • major therapeutic benefits could be obtained by means of intracellular delivery of toxic peptides such as ricin, diphtheria toxin, or cobra venom factor in order to eliminate diseased or malignant cells.
  • Transfection of cells has been carried out by various methods, comprising calcium phosphate precipitation, or DE
  • cationic lipid technology based on a discovery that a positively charged synthetic cationic lipid, N- [ 1- ( 2 # 3-dioleyloxy)propyl]-N,N,N- trimethylammonium chloride (DOTMA) , in the form of liposomes, or small vesicles, could interact spontaneously with DNA, which is negatively charged, or anionic, to form lipid-DNA complexes having a net positive charge and capable of fusing with the negatively charged cell membranes of tissue culture cells, to achieve both uptake and expression of the DNA (Feigner, P.L.
  • DOTMA a positively charged synthetic cationic lipid
  • DNA which is negatively charged, or anionic
  • the Lipofectin w reagent (Bethesda Research Laboratories, Gaithersburg, Maryland) , an effective commercial cationic lipid reagent for the delivery of highly anionic polynucleotides into living tissue culture cells, comprises positively charged DOTMA liposomes.
  • DOTMA analogue 1,2-bis(oleoyloxy)-3-(trimethylammonio)- propane (DOTAP) has been successfully used in combination with a phospholipid to form DNA-complexing vesicles
  • Active Molecules filed April 19th, 1990 by Feigner et al., discloses cationic lipids which are more effective and less toxic than presently known cationic lipids for transfection.
  • Cationic lipid methodology is presently preferred over other methods; it is more convenient and efficient than calcium phosphate, DEAE dextran or electroporation methods.
  • it can only be applied to polyanionic molecules, such as DNA.
  • polyanionic molecules such as DNA.
  • bioactive substances of interest are as negatively charged as polynucleotides; therefore, the extension of cationic lipid methodology to the delivery of other molecules, for example, many proteins, is not feasible.
  • Transport of proteins is among the most selective and specific of transmembrane processes. Such processes involve protein-protein recognition (signal sequences) , frequently require ATP, or are enzymatically driven. Viruses have proteins capable of accessing these processes successfully, thus allowing viral infections of cells to occur. The viral method of intracellular penetration has been exploited to develop a methodology which uses viral "chimerasomes" or proteoliposomes for intracellular protein delivery. Viral chimerasomes are lipid vesicles containing molecules intended for intracellular delivery together with two critical proteins isolated from viruses. One of these proteins is a cell surface recognition protein which promotes attachment of lipid vesicles with the cell surface.
  • a viral fusion protein which facilitates the introduction of the viral contents into the cell cytoplasm.
  • the chimerasome methodology is broadly applicable to the delivery of proteins as well as polynucleotides.
  • it is more expensive and difficult to manufacture than cationic lipid reagents because it involves isolation and purification of components of potentially infectious virus particles. Stability of the proteins and lipids in the chi erosome reagent is a potential problem, and it is also somewhat less convenient to use than the cationic lipids.
  • polynucleotide molecules are first treated with DOTMA to produce a positively charged complex.
  • This complex is then treated with negatively charged liposomes to form a double coated, negatively charged complex.
  • the negatively charged coat may comprise a coupling reagent.
  • Receptor specific agents such as antibodies, lectins, and other ligands may then be attached to the negatively charged outer layer through the coupling reagent.
  • the overall negative charge of the double-coated complex prohibits its non-specific interaction with the negatively charged membranes of cells while allowing relatively specific interaction through the binding of integral ligands.
  • the defect of this approach like that of the chimerosome methodology, is its relative complexity, and its requirement for specific binding reagents; further, its application, like that of the parent cationic lipid methodology, is limited to polyanionic materials, that is. polynucleotides and negatively charged proteins.
  • the intracellular delivery systems disclosed herein exploit self-assembling processes found in natural systems to join together molecular subcomplexes through physical mechanisms of ionic attraction and repulsion and hydrophobic-hydrophilic interactions.
  • the most pertinent example of such systems in nature is the simple assemblies of lipids and proteins to form cell membranes. The physical forces holding such structures together are relatively weak and non-specific, and this, among other factors, allows the assembly to proceed spontaneously and rapidly.
  • the following figures show the intracellular delivery of proteins into tissue cells in culture according to the methods of the invention. Each figure consists of a pair of photomicrographs produced by (a) phase contrast and (b) fluorescence microscopy.
  • Figure 1 shows intracytoplasmic delivery of fluoresceinated dextran (MW 4000) , into NIH-3T3 cells.
  • Figure 2 shows intracytoplasmic delivery of fluoresceinated dextran (MW 4000) into HELA cells.
  • Figure 3 shows the intracellular delivery of Rhoda ine-PE into NIH-3T3 cells.
  • Figure 4 shows the delivery of a low molecular weight dye, 6CF, into NIH-3T3 cells.
  • Figure 5 shows the delivery of Rhodamine-phalloidin into NIH-3T3 cells.
  • the invention provides a method for making a vehicle for administering a bioactive substance to a cell, comprising the steps of providing the substance in a first lipid vesicle which vesicle comprise at least one negatively charged lipid and is negatively charged; and combining this first lipid vesicle with second lipid vesicles comprising at least one cationic lipid and having a net positive charge, whereby the first negatively charged vesicles are coated with at least one positively charged lipid vesicle.
  • the bioactive substance can be encapsulated in the vesicle by containment in the aqueous core; alternatively, if the bioactive substance is lipophilic or has a lipophilic or amphipathic region, it may be incorporated by means of that lipophilic structure into the lipid wall of the vesicle.
  • a method for making a vehicle for administering a bioactive substance to a cell comprising the steps of forming a first complex comprising a positively charged macromolecule and at least one lipid vesicle, wherein the lipid vesicle comprise a negatively charged lipid and has a net negative charge; and contacting the first complex with lipid vesicles comprising at least one cationic lipid and having a net positive charge to form a second complex.
  • a method for administering a bioactive substance to a cell comprising the steps of providing the substance in a negatively charged lipid vesicle; and administering the negatively charged lipid vesicle to a cell together with a lipid vesicle comprising at least one cationic lipid and having a net positive charge.
  • a method for administering a bioactive substance to a cell comprising the steps of preparing a complex comprising said bioactive substance and a negatively charged lipid vesicle; and administering the complex to a cell together with a lipid vesicle comprising at least one cationic lipid and having a net positive charge.
  • the methods of administering bioactive substances to a cell can be applied in vivo as well as in vi tro .
  • the invention also provides a composition of matter, comprising a bioactive substance contained in a negatively charged lipid vesicle and lipid vesicles, comprising at least one cationic lipid species and having a net positive charge, at least partially covering the negatively charged lipid vesicle.
  • the composition has a net positive charge.
  • the bioactive substance of the composition can be a biological macromolecule, such as, for example, a polynucleotide, a protein or polypeptide.
  • the bioactive substance may also be a drug, and the drug may be a small organic molecule.
  • a bioactive substance is encapsulated within a negatively charged lipid vesicle.
  • the bioactive substance may also be lipophilic or comprise a lipophilic region, and is thereby incorporated among the lipids of the lipid vesicle.
  • the invention also provides a kit, comprising materials, supplies, reagents, and instructions for preparing self-assembling lipid complexes for the delivery of a bioactive substance to a cell, either in vi tro or in vivo , comprising at least one vessel containing at least one negatively charged amphipathic lipid species; and a quantity of a lipid substance, the substance comprising at least one positively charged amphipathic lipid species.
  • the kit can further comprise at least one hydrating buffer solution and an instruction leaflet.
  • suitable positively charged lipid species can be known or unknown cationic lipids, such as, for example DOTAP orDOTMA or can have the structure of any of the novel cationic lipids described herein.
  • the present invention provides vehicles, comprising self-assembling lipid complexes, capable of delivering bioactive substances, particularly polynucleotides, oligonucleotides, proteins, peptides or drugs into living cells in a convenient and efficient manner.
  • lipid vesicles comprising lipid species having either a negative or positive charge, are combined with the substance to be delivered in a properly ordered sequence, forming self-assembling lipid complexes containing the substance to be delivered and having a positively charged outer lipid layer.
  • the invention like cationic lipid technology, takes advantage of the natural fusion properties of a positively charged lipid to promote passage of bioactive substances associated with that lipid across the negatively charged cell membrane.
  • the invention provides a prior association of the bioactive substance with negatively charged lipids to form primary complexes having a net negative charge. This primary complex promotes a subsequent secondary complex formation with the liposomes comprising the cationic lipids that facilitate membrane transport. In this way the invention extends cationic lipid methodology to all bioactive agents intended for intracellular delivery, regardless of size or net charge.
  • Vehicles for intracellular delivery according to the invention comprise a primary negatively charged lipid complex in association with the bioactive substance to be delivered and a secondary complex, having a net positive charge and comprising the primary negative complex in association with positively charged liposomes.
  • the invention provides two general strategies for assembling the primary negatively charged lipid complex comprising the bioactive substance.
  • the bioactive substance to be delivered is entrapped within a lipid vesicle or liposome comprising negatively charged lipids and having a net negative charge.
  • the entrapment can occur, according to one embodiment of the invention, as an incorporation of a lipophilic bioactive substance, or such a substance having a lipophilic region, into the wall of the lipid vesicle.
  • the entrapment can occur as an encapsulation of hydrophilic bioactive substance into the aqueous core of the lipid vesicle.
  • hydrophilic bioactive substance For both lipophilic and hydrophilic substances, the uptake into the negatively charged liposome occurs during the process of liposome formation, typically the hydration of a lipid film, disclosed herein, and illustrated by examples. Examples 3, 5, and 6 disclose the encapsulation of substances within negatively charged liposomes, and Example 4 discloses the incorporation of a lipid derivative into the liposome wall.
  • the bioactive substance is contacted with negatively charged liposomes, and these liposomes then at least partially enclose the substance to provide a primary lipid complex having negatively charged regions.
  • the bioactive substance has a net positive charge and interacts with the negative liposomes by means of charge attraction; alternatively, the bioactive substance can be neutral or negatively charged but comprise lipophilic regions that promote its association with the liposomes of the complex through hydrophobic or lipophilic interactions.
  • the bioactive substance can comprise amphipathic peptide helices, for example of the type disclosed by Segrist, U.S. Patent No.
  • the negatively charged liposomes used to form the complex may contain the same or other bioactive substances, either encapsulated or incorporated, as disclosed above.
  • the second element of the intracellular delivery system of the invention comprises liposomes formed from positively charged cationic lipids and having a net positive charge.
  • the primary complex comprising either the negatively charged liposomes or the negatively charged lipid complex, the positively charged liposomes spontaneously form complexes having a net positive surface charge.
  • the positively charged assembled complexes when placed in contact with cells, will spontaneously attach to cell surfaces and interact directly with the plasma membrane, according to conventional cationic lipid methodology. Delivery of the bioactive cargo into the cell cytoplasm can occur by various mechanisms.
  • the lipids of the complex may either directly fuse with the plasma membrane and discharge the entrapped substance intracellularly; alternatively, it may be phagocytized and interact with other internal membranes or the membranes of the phagocytic compartment itself.
  • All of these assemblies comprising the encapsulation or incorporation of substances within lipid vesicles, the association of substances with positively or negatively charged lipids or vesicles thereof, the association of positively and negatively charged lipids and vesicles thereof, and even the attachment of the vesicles to the target cell surface and fusion directly with cell membranes, occur spontaneously as the result of repulsions and attractions of ionic charges and hydrophobic-hydrophilic interactions.
  • the negative and positively charged lipid complexes form almost instantaneously, and accordingly the primary negatively charged complexes can be contacted with the positively charged liposomal preparation and target cells simultaneously.
  • the primary negative complexes and positively charged liposomes may be mixed to form secondary complexes before contact with the target cells.
  • prior complex formation is preferred, while for in vi tro applications, the positively charged empty liposomes are conveniently added to a cell culture at the same time as the negatively charged liposome-bioactive agent complexes, thus avoiding a separate mixing step.
  • the lipid reagents of the invention may comprise lipid mixtures similar to that of the physiological cell membrane, comprising phospholipids as primary components.
  • the lipid reagents can further comprise any of the conventional synthetic or natural liposome materials, including phospholipids from natural plant or animal sources such as phosphatidylcholine . phosphatidylethanolamine, sphingomyelin, phosphatidylserine, or phosphoinositol.
  • Synthetic phospholipids that may also be used include, but are not limited to, dimyristoylphosphatidylchol ine , dioleoylphosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphatidylcholine and the corresponding synthetic pho s ph at i dy 1 e th ano 1 ami ne s and phosphatidylglycerols.
  • Other additives such as cholesterol, glycolipids, fatty acids, sphingolipids, or gangliosides can also be used, as is conventionally known for the preparation of liposomes.
  • the positively and negatively charged lipid vesicles used in the methods of the invention are typically prepared as appropriate from a mixture of either cationic lipids or negatively charged lipids, neutral lipids and cholesterol or a similar sterol.
  • Neutral lipids can be phosphatidylcholine, phosphatidyl ethanolamine, similar phospholipid analogs, or mixtures of these, as well as monoglycerides, diglycerides and trigly ⁇ erides.
  • the negatively charged lipid reagents of the invention are those comprising at least one lipid species having a net negative charge at physiological pH or combinations of these. Suitable lipid species comprise phosphatidyl glycerol and phosphatidic acid or a similar phospholipid analog.
  • the positively charged lipid reagents of the invention are those comprising at least one cationic lipid species having a net positive charge at physiological pH.
  • Suitable lipid species comprise known cationic lipids, such as l,2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP) or N-( ⁇ , ⁇ -l-dialkoxy)-alk-l-yl-N, N, N-trisubstituted ammonium surfactants, such as DOTMA, or complex cationic lipids having similar structures and properties or mixtures of these.
  • Particularly preferred cationic lipids are those disclosed in a co-pending U.S. Application entitled Cationic Lipids for Intracellular Delivery of Biologically Active Molecules filed April 19th, 1990 by Feigner et al., which is hereby incorporated by reference.
  • Y 1 and Y- are the same or different and are -0-CH2- -
  • R 1 and R 2 are the same or different and are H, or C-_ to C2 3 alkyl or alkenyl;
  • R 3 and R 4 are the same or different and are Ci to C24 alkyl, or H;
  • R 5 is Ci to C 24 alkyl straight chain or branched chain
  • R 6 is -C(0)-(CH2)m _ NH-, a diaminocarboxylic acid which is alkyl, aryl, or aralkyl, or -C(O)-(CH 2 )m ⁇ NH ⁇ linked to said diaminocarboxylic acid, or is absent;
  • R 7 is H, spermine, spermidine, a histone, or a protein with
  • R 7 is an L- or D-alpha amino acids having a positively charged group on the side chain, said amino acids comprising arginine, histidine, lysine or ornithine or derivatives thereof, or the same amino acids wherein the amine of the R7 moiety is quaternized with R 3 , R 4 or R 5 groups; or
  • R 7 is a polypeptide selected from the group consisting of
  • L- or D-alpha amino acids wherein at least one of the amino acids residues comprises arginine, histidine, lysine, ornithine, or derivatives thereof; n is 1 to 8; m is 1 to 18; and
  • X is a non-toxic anion
  • the positively charged liposomes of the invention comprise the cationic lipids l,2-dioleoyl-3- dimethylaminopropyl-j8-hydroxyethylammonium or 1-0-oleyl-, 2-oleoyl-3-dimethylaminopropyl-0-hydroxyethylammonium.
  • ester or ester derivatives of the known cationic lipids having a formula as set forth below are also preferred.
  • Y 1 and Y 2 are different and are either -0-CH2-, -O-C(O)-, or OH;
  • R 1 and R 2 are individually absent or are Ci to C23 alkyl or alkenyl; R 3 , R 4 and R 5 are the same or different and are H, C ⁇ to
  • R 3 , R 4 , and R 5 are taken together to form quinuclidino, piperidino, pyrrolidino, or morpholino; n is 1 to 22; and X is a non-toxic anion.
  • Non-toxic anions described herein may be those of pharmaceutically non-toxic acids including inorganic acids and organic acids. Such acids include hydrochloric, hydrobromic, sulfuric, phosphoric, acetic, benzoic, citric, glutamic, lactic acid and the like.
  • acids include hydrochloric, hydrobromic, sulfuric, phosphoric, acetic, benzoic, citric, glutamic, lactic acid and the like.
  • the cationic lipid in a lipid formulation for preparing positively charged lipid vesicles, can be present at a concentration of between about 0.1 mole% and 100 mole%, preferably 5 to 95 mole%, and most preferably between 20 and 80 mole%. In a formulation for preparing negatively charged lipid vesicles, the negatively charged lipid can be present at a concentration between about 0.1 and 100 mole%, preferably 1 to 90 mole%, and most preferably 3 to 50 mole%.
  • the alternatively charged lipid can be present at between about 0 to 49 mole% and preferably 0 to 40 mole%.
  • the neutral lipid can be present in positively or negatively charged lipid vesicles in a concentration of between about 0 and 99.9 mole %, preferably 5 to 95 mole%, and most preferably 20 to 80 mole%. Cholesterol or a similar sterol can be present at 0 to 80 mole %, and preferably 0 to 50 mole%.
  • the lipid reagents may be prepared and stored as empty liposomes in aqueous solution, or may be stored as dried lipid, for example as a lipid film, after formulation to be later used as encapsulating reagents for selected bioactive substances. Liposome Formation
  • the lipid formulations of the invention comprising at least one amphipathic lipid, such as a phospholipid, spontaneously assemble to form primary liposomes, heterogeneous in size and structure, in aqueous solution. Therefore the term lipid reagent, lipid vesicles, and liposomes are used interchangeably in describing formulations.
  • the lipid reagents either negatively or positively charged, are prepared as liposomes or lipid vesicles, according to Example 1.
  • the component lipids are dissolved in a solvent such as chloroform and the mixture evaporated to dryness as a film on the inner surface of a glass vessel.
  • the amphipathic lipid molecules On suspension in an aqueous solvent, the amphipathic lipid molecules assemble themselves into primary liposomes. If other molecules are present in the aqueous solvent, such as, for example, a bioactive substance, these will be captured within the liposomes, as demonstrated in Examples 3 and 5.
  • some lipophilic bioactive substances intended for entrapment can be dissolved or suspended in the organic solvent that is used to dissolve the lipids of the liposome formulation. After evaporation of the lipid film and hydration to form primary liposomes, the substances thus dissolved are typically incorporated into the bilayer of those liposomes, rather than being encapsulated into the aqueous interior. Otherwise, if there is no solute in the hydration buffer, empty liposomes will be formed, as demonstrated in Examples 1 and 2.
  • the primary liposomes are preferably processed by the freeze-thaw and extrusion processes, as provided by Example 2. These primary liposomes are reduced to a selected mean diameter by means of the freeze-thaw procedure referred to above.
  • the cationic lipids of the invention are formed into vesicles of uniform size prior to transfection procedures, according to methods for vesicle production published in the literature and knov/n to those in the art, for example, the sonication of spontaneously formed liposomes comprised of the lipids in aqueous solution described by Feigner, P.L.
  • Suitable conventional methods of preparation include, but are not limited to, those disclosed by Bangham, A. et al., J. Mol. Biol. 23: 238-252 (1965); Olson, F. et al., Biochim. Biophys. Acta 557: 9-23 (1979), Szoka F. et al., Proc. Natl. Acad. Sci. USA 75: 4194-4198 (1978), Mayhew, E. et al. Biochim. Biophys. Acta 775: 169-175 (1984), Kim, S. et al. Biochim. Biophys. Acta 728:339-348) , and Mayer, L. et al., Biochim. Biophys. Acta 858:161-168 (1986).
  • the negative and positive lipid reagents of €he invention comprising at least one amphipathic lipid species, such as a phospholipid, are prepared using the freeze-thaw-extrusion procedure indicated in Examples 1 and 2.
  • the component lipids are dissolved in a solvent such as chloroform and the mixture evaporated to dryness as a film on the inner surface of a glass vessel.
  • the amphipathic lipid molecules assemble themselves into primary liposomes. If other molecules are present in the aqueous solvent, such as, for example, a bioactive substance, these will be captured within the liposomes, as indicated in Examples 3, 4, 5 and 6. Otherwise, empty liposomes will be formed, as in Example 1.
  • These primary liposomes are reduced to a selected mean diameter by means of the freeze-thaw procedure described in Example 2.
  • the bioactive cargo of the lipid delivery system is entrapped into negatively charged liposomes.
  • the lipid derivatives of agents disclosed above for example, the lipid derivatives of antiviral nucleoside ⁇ such as phosphatidylazidc nymidine or (3'-azido-3 , -deoxy)thymidine- 5'-diphospho-3-diacylglycerol, can be entrapped by direct incorporation into the wall of the lipid vesicle during the hydration of a lipid film comprising these lipid derivatives in the liposome formulation.
  • Other agents can be encapsulated within the aqueous space of the liposome according to conventional liposome forming methodology, as follows.
  • Substances intended for intracellular delivery can be encapsulated into negatively or positively charged liposomes by any one of a number of standard procedures. For example, each of the lipid components comprising the liposomal formulation is dissolved together into a co- miscible organic solvent. If a lipid derivative of an agent to be delivered is to be incorporated into the liposome, a selected quantity of that agent is added to the other lipid components at this time. The solvent is evaporated and the vessel containing the residual lipid film is evacuated overnight to remove solvent traces.
  • hydration solution is next added to the dried lipid film to form primary liposomes. If hydration buffer alone is added to a film of lipid components alone, empty liposomes will form. If hydration buffer alone is added to a lipid film comprising lipid derivatives of bioactive agent, the primary liposomes will comprise that agent incorporated into the vesicle walls. If hydration buffer having a bioactive dissolved therein is added to a lipid film comprising only lipids, the primary liposomes that form will comprise that agent trapped in the interior aqueous compartment of the liposomes. Permutations of the processes, resulting in liposomes comprising bioactive agents both incorporated into the walls of the vesicle as well as entrapped therein are possible and are within the contemplation of the invention.
  • the hydration solution can be any biologically compatible buffer solution comprising isotonic saline or phosphate buffered saline, or low ionic strength buffers comprising 5% sorbitol or 10% sucrose. Such buffers are well known to those skilled in the art.
  • concentration of the bioactive substance in the hydration buffer which is intended for intracellular delivery can vary widely depending on the substance or the application; this concentration can be between 1 picogram/ml and 500 mg/ml.
  • the resulting liposome suspension can be further emulsified by any one of a number of procedures; for example, the sample can be forced through Nuclepore'" membranes to produce vesicles of a size comparable to the pore size of the membranes.
  • Encapsulations are described in Examples 3, 4, and 6 for the encapsulation of FITC dextran, 6-carboxy-fluorescein, and phalladion in the negatively charges lipid formulation of 4.9/2.1/3 DOPC/DOPG/cholesterol, followed by freeze- thaw and extrusion to achieve a uniform sized liposomal preparation. Any of the substance remaining unencapsulated can be removed, if desired, by a process such as gel filtration chromatography.
  • the procedure for delivering materials into cells according to the method of the invention comprises a strategy of presenting the material to the cell in association with positively and negatively charged lipid vesicles, described as follows, and illustrated by the accompanying Examples.
  • the resulting liposomes or complexes are added directly to cells, for example in an in vi tro application, in a suitable biologically compatible medium, together with positively charged lipid vesicles.
  • concentration of lipid with which the cells are contacted varies widely depending on the application, but is between 1 ⁇ olar and
  • the positively charged lipid vesicles attach spontaneously to the target cell surface, fuse with the cellular membranes and deliver the contents of the liposome into the cell cytopiasm.
  • the substance intended for intracellular delivery is first encapsulated into negatively charged liposomes, or complexed with empty negatively charged liposomes, according to the procedures described previously. These primary negative complexes are then exposed to a quantity of positively charged lipid vesicles. Immediately upon mixing the two solutions containing the oppositely charged lipid vesicles, complexes spontaneously form comprising negatively and positively charged vesicles.
  • the quantity of positively charged vesicles added to the encapsulating negatively charged vesicles should be sufficient to encourage attachment of the complexes to the target cell surface. Theoretically, this quantity should be sufficient to provide the complexes with a net positive charge, with the number of positive charges contributed by the positively charged liposomes in excess over the number of negative charges contributed by the negatively charged liposomes; however, such an excess of positively charged elements is not always required to encourage sufficient attachment to the target cell surface to achieve intracellular delivery. Accordingly, the ratio of positive to negative charges in the final complexes may be from about 100:1 to 0.1:1 and preferably 20:1 to 0.2:1.
  • the present invention also contemplates the use of liposome complexes comprising positively charged liposomes but having a net overall negative charge for the intracellular delivery of negatively charged polynucleotides.
  • liposome complexes comprising positively charged liposomes but having a net overall negative charge for the intracellular delivery of negatively charged polynucleotides.
  • These complexes are prepared as follows: Polynucleotides in solution are mixed with a sufficient quantity of positively char:sd liposomes so as to form a complex which reduces the anionic characteristics of the polynucleotide by a sufficient amount as described below.
  • Negatively charged elements in a polynucleotide solution can be readily quantified using the extinction coefficient for the nucleotide monomer which bears one negative charge per monomer.
  • the concentration of polynucleotide in solution can be from about 0.01 ⁇ g/ml to 50 mg/ml, preferably from 1 ⁇ g/ml to 10 mg/ml and most preferably from 10 ⁇ g/ml to 1 mg/ml.
  • the concentration of positively charged lipid vesicles can range from between 0.1 ⁇ g/ml to 100 mg/ml, preferably from 1 ⁇ g/ml to 100 mg/ml, and most preferably from 10 ⁇ g/ml to 50 mg/ml.
  • the solutions may be mixed together from those having a low ionic strength buffer, that is having an ionic strength less than that of 25 mM sodium chloride. Sorbitol, sucrose or glucose can be used to render a low ionic strength buffer isotonic. Adsorption of the polyanionic polynucleotides to the cationic vesicles reduces the negative charge character of the polynucleotides. In theory, the quantity of charges contributed by the vesicles should exceed the number of negative charges contributed by the polynucleotide, although this condition may not be an absolute requirement for every application.
  • the ratio of positive to negative charges in the polynucleotide/cationic lipid complexes may be from about 100:1 to 0.1:1 and preferably 20:1 to 0.2:1.
  • the entire polynucleotide can be incorporated by self-assembly into a lipid complex that can accomplish intracellular delivery without a loss in efficiency caused by incomplete nucleotide encapsulation.
  • the quantity of negatively charged liposomes added should exceed the net positive charge contributed by the complex of positively charged lipid and polynucleotide, although in practice this level of negatively charged elements may not be required for every application. Accordingly, the ratio of positive to negative charges in the polynucleotide/cationic lipid complexes may be 100:1 to 0.1:1 and more preferably 20:1 to 0.2:1.
  • the negatively charged complexes produced above, containing polynucleotide and positively charged lipids, are analogous to the primary negatively charged lipid complexes disclosed for other bioactive substances, and can be delivered in vivo or in vi tro to tissue culture cells in a manner similar to that described above for those primary complexes.
  • Fluoresceinated dextran which is available in different molecular sizes from 4,000 to 70,000 MW and having a net positive, neutral or negative charge. Delivery of dextran into the cell was quantitated by total cell associated fluorescence as determined by a spectrophotometer. The distribution of fluorescence was determined by fluorescence microscopy. A thin band of fluorescence around the cell was read to indicate cell-associated vesicles; bright spots around the nucleus to indicate lysosomal uptake; and diffuse cytoplasmic fluorescence to indicate cytoplasmic delivery. Cytoplasmic delivery is considered the most effective.
  • Fluoresceinated phalloidin This 7-amino acid cyclic peptide binds specifically to intracellular actin. Functional delivery of this substance is indicated by the filamentous intracellular staining pattern characteristic of actin filaments seen on examination of the cells by fluorescence microscopy.
  • Anti-actin antibody This antibody will bind to actin when delivered intracellularly. Treated cells were fixed, permeabilized and counter-stained with a fluorescent antibody against the anti-actin antibody. As in the case of fluoresceinated phalloidin, effective delivery is indicated by the characteristic actin pattern seen on fluorescence microscopy. Cell viability for all experiments was determined by trypan blue staining.
  • the self-assembling delivery systems of the invention can be advantageously formed using any cationic lipid, whether those previously known, for example, DOTMA or DOTAP, or novel cationic lipids, such as those described above and also disclosed in co-pending U.S. Application of Feigner et al., filed April 19, 1990, to deliver substances intracellularly either in vitro or in vivo.
  • Those cationic lipids described above having metabolizable ester bonds are preferred for in vi vo use.
  • vi tro applications include the intracellular delivery to any cell grown in culture, comprising cells of any species, whether plant or animal, vertebrate or invertebrate, and of any tissue or type.
  • Contemplated uses comprise transfection procedures corresponding to those presently known. Accordingly, the strategies of the invention can be used to facilitate the intracellular delivery of DNA or mRNA sequences coding for therapeutically active polypeptides, as described in detail in U.S. Patent Applications Serial Nos. 326,305 and 467,881 which are hereby incorporated by reference.
  • the self- assembling delivery methods described herein are particularly preferred for the delivery of the expressed gene product or protein itself.
  • the self-assembling delivery of proteins can provide therapy for genetic disease by supplying deficient or absent gene products to treat any genetic disease in which the defective gene or its product has been identified, such as Duchenne's dystrophy (Kunkel, L. and Hoffman, E. Brit. Med. Bull. 45(3) :630-643 (1989) or cystic fibrosis (Goodfellow, P. Nature. 341(6238) :102-3 (Sept. 14, 1989).
  • the self-assembling delivery systems described above can also provide immunizing polypeptides to the cell either by delivering a polynucleotide coding for the immunogen, or the immunogen itself.
  • Other therapeutically important polynucleotides suitable for self-assembling delivery comprise anti-sense polynucleotide sequences, useful in eliminating or reducing the production of a gene product, as described by Ts'o, P. et al. Annals New York Acad. Sci. 570:220-241 (1987).
  • the delivery by means of the methods disclosed, ribozymes, or catalytic RNA species, either of the "hairpin” type as described by Ha pel et al. Nucleic Acids Research 18(2) :299-304 (1990); or the "hammerhead” type described by Cech, T. and Bass, B. Annual Rev. Biochem. 55:599-629 (1986) .
  • Particularly preferred within the contemplated uses of the invention are deliveries of either an anti-sense polynucleotide or ribozy e as described above, and having as its target the rev site if the HIV genome (Scientific American, October, 1988, pp. 56-57).
  • Matsukura, M. et al. Proc. Natl. Acad. Sci. 86:4244-4248 (1989) describe a 28-mer phosphorothioate compound anti-HIV (anti-rev transactivator) specific for the site.
  • the transfection procedures described above may be applied by direct injection of self-assembled lipid complexes, comprising DNA, RNA or proteins into cells of an animal in vi vo .
  • the above therapies can be alternatively carried out by in vi tro transfection of some of the cells of an animal using self-assembling delivery methods, and re- introduction of the cells into the animal.
  • the ability to transfect cells at high efficiency with self-assembling delivery thus provides an alternate method for immunization.
  • the gene for an antigen is introduced into cells which have been removed from an animal by self- assembling delivery methods.
  • the trans-fected cells, now expressing the antigen are reinjected into the animal where the immune system can now respond to the (now) endogenous antigen.
  • the process can possibly be enhanced by co-injection of either an adjuvant or lymphokines to further stimulate the lymphoid cells.
  • compositions and methods of the present invention can be most advantageously used in the periodic supplying of endogenous or exogenous macromolecules, particularly proteins, to a cell. They are therefore particularly suitable for use in transient therapies which requires treatment with proteins, particularly in cells unable to carry out translation of messenger RNA.
  • transient therapeutic uses of self- assembling delivery methods herein disclosed include the liposomal delivery of nucleotide analogues having an antiviral effect, such as dideoxynucleosides, didehydronucleosides, nucleoside analogues having halogen substituted and azido-substituted ribose moieties, such as 3* * -azido-3 , deoxythymidine (AZT) , or nucleoside analogues such as acyclovir or gancyclovir (DHPG) .
  • liposomal delivery of nucleotide analogues having an antiviral effect such as dideoxynucleosides, didehydronucleosides, nucleoside analogues having halogen substituted and azido-substituted ribose moieties, such as 3* * -azido-3 , deoxythymidine (AZT) , or nucle
  • peptides comprising physiologic species such as interleukin-2, tumor necrosis factor, tissue plasminogen activator, factor VIII:C, erythropoietin, growth factors such a ⁇ epidermal growth factor, neural growth factor, and hormones such as tissue insulin, calcitonin, and human growth hormone as well as toxic peptides such as ricin, diphtheria toxin, or cobra venom factor, capable of eliminating diseased or malignant cells.
  • physiologic species such as interleukin-2, tumor necrosis factor, tissue plasminogen activator, factor VIII:C, erythropoietin
  • growth factors such a ⁇ epidermal growth factor, neural growth factor, and hormones such as tissue insulin, calcitonin, and human growth hormone as well as toxic peptides such as ricin, diphtheria toxin, or cobra venom factor, capable of eliminating diseased or malignant cells.
  • toxic peptides such as ricin, diph
  • formulations preferably comprise lipid derivatives of the antiviral agents, particularly the phosphatidylglycerol derivatives as disclosed in U.S. Applications Serial Nos. 216,412, 319,485 and 373,088.
  • the effect of the self-assembled lipid delivery system is to facilitate the penetration of the active antiviral agent through the stratum corneum of the dermis.
  • Self-assembling systems comprising lipid-encapsulated bioactive agents may accordingly be used for intracellular delivery either in vi vo or in vi tro .
  • neutral or positively charged bioactive molecules are encapsulated in a negatively charged lipid reagent and added to a washed cell culture together with a volume of a suspension of liposomes comprising the positively charged lipid reagent, as indicated in Example 8.
  • corresponding molecules are encapsulated with negatively charged lipid reagent and then coated with the liposomes of the positively charged lipid reagent before administration.
  • Lipids used l,2-Bis(oleoyloxy)-3-(trimethylammonio) propane, DOTAP; dioleoyl phosphatidylethanolamine, DOPE.
  • Lipids used dioleoylphosphatidylcholine, DOPC; dioleoylphosphatidylglycerol, DOPG; cholesterol Quantities of 4.67 mg DOPC, 4.06 mg DOPG and 2.9 mg cholesterol comprising 25 uMoles of total lipid, in a molar ratio of 4.9/2.1/3, DOPC/DOPG/cholesterol, were combined in 1 ml of chloroform. The mixture was dried on a Rotavap apparatus and the flask evacuated overnight on a vacuum pump.
  • the lipid film was suspended at 20"C for 1 hour using a rotary shaker with l ml tris buffer (20 mM pH 7.5) and the suspension adjusted to 290 mOsm by adding crystalline NaCl.
  • the sample was frozen in a dry ice/isopropanol bath and thawed in a 30°C bath.
  • the sample was then extruded under 200-750 psi using two stacked 0.2 micron polycarbonate filter membranes (Nuclepore”) in a 10 ml volume "Extruder” (Lipex Biomembranes Inc. , Vancouver, Canada) .
  • the freeze/thaw and extrusion was repeated two more times.
  • the final product was extruded two more times.
  • a quantity of 200 mg FITC Dextran (Sigma Chemical Co.) was dissolved in 1.5 ml 20 mM Tris buffer (pH 7.5) and the solution adjusted to 290 mOsm with NaCl.
  • a lipid film comprising 25 uMoles of total lipid, in a molar ratio of 4.9/2.1/3, DOPC/DOPG/Cholesterol, was prepared as in Example 2.
  • the lipid film at 20 ⁇ C was suspended in 1.5 ml of the FITC Dextran solution for 1 hour using a rotary shaker.
  • the sample was frozen in a dry ice/isopropanol bath and thawed in a 30°C bath, then extruded under 200-750 PSI through stacked 0.2 micron polycarbonate filter membranes (Nuclepore'") in a 10 ml volume "Extruder” (Lipex Biomembranes Inc. , Vancouver, Canada) .
  • the freeze/thaw and extrusion was repeated two more times.
  • the final product was extruded two more times.
  • the sample was applied to a Sephadex G-75 column equilibrated with the 20 mM tris buffer (adjusted to 290 mOsm with NaCl) and the void volume, which contains the liposome encapsulated FITC dextran, collected.
  • a quantity of 22 mg egg phosphatidylcholine, 9.3 mg egg phosphatidylglycerol, 7.7 mg cholesterol and 1 mg of rhodamine-phosphatidylethanolamine were dissolved into 1 ml of chloroform.
  • the lipid film was suspended at 20"C for 1 hour using a rotary shaker with Dulbecco's phosphate buffered saline (PBS) pH 7.4.
  • PBS Dulbecco's phosphate buffered saline
  • the sample was frozen in a dry ice/isopropanol bath and thawed in a 30 ⁇ C bath.
  • the sample was extruded under 200-750 psi using two stacked 0.1 micron polycarbonate filter membranes (Nuclepore*) in a 10 ml volume "Extruder” (Lipex Biomembranes Inc., Vancouver,
  • EXAMPLE 5 Encapsulation of 6 carboxy-fluorescein in Negatively Charged Liposomes
  • 6CF carboxy-fluorescein
  • phosphate buffer pH 7.4
  • IN NaOH phosphate buffer
  • This solution contained 8.8 mg/ml 6CF (23.4 mM) .
  • a quantity of 9.34 mg DOPC, 8.12 mg DOPG and 5.8 mg cholesterol was placed into 2 ml of chloroform, comprising 50 ⁇ M of total lipid in a ratio of 4.9/2.1/3 DOPC/DOPG/Cholesterol.
  • the lipid film was suspended at 20 ⁇ C for 1 hour using a rotary shaker with 0.75 ml of the 6CF solution.
  • the sample was frozen in a dry ice/isopropanol bath and thawed in a 30"C bath.
  • the sample was then extruded under 200-750 psi using two stacked 0.2 micron polycarbonate filter membranes (Nuclepore) in a 10 ml volume "Extruder” (Lipex Biomembranes Inc., Vancouver, Canada) .
  • the freeze/thaw and extrusion was repeated two more times.
  • the final product was extruded two more times.
  • the sample was frozen in a dry ice isopropanol bath and thawed at room temperature.
  • the phalloidin peptide was dissolved into 0.5 ml of Tris buffer (20 mM pH 7.5) and adjusted to 290 mOsm) .
  • a quantity of 4.67 mg DOPC, 4.06 mg DOPG and 2.9 mg cholesterol was taken up into 1 ml of chloroform. This results in 25 uMoles of total lipid, in a molar ratio of 4.9/2.1/1/3, DOPC/DOPG/Cholesterol.
  • the lipid film was suspended at 20° C for 1 hour using a rotary shaker with 1.5 ml of the phalladion solution.
  • the sample was frozen in a dry ice/isopropanol bath and thawed in a 30*C water bath.
  • the sample was then extruded under 200-750 psi using two stacked 0.2 micron polycarbonate filter membranes (Nuclepore"') in a 10 ml volume “Extruder” (Lipex Biome branes Inc. , Vancouver, Canada) .
  • the freeze/thaw and extrusion was repeated two more times.
  • the dried lipid film was suspended in 3 ml of the phalloidin solution.
  • the sample was frozen in a dry ice/isopropanol bath and thawed at room temperature. It was then extruded though 0.2 micron nucleopore filter membranes.
  • the freeze/thaw and extrusion was repeated two more times.
  • the final product was extruded two more times.
  • the sample was then applied to a Sephadex G-75 column equilibrated with the 20 mM tris buffer (adjusted to 290 mOsm with NaCl) and the void volume, containing the liposome encapsulated phalladion collected.
  • PBS Dulbecco s phosphate buffered saline without calcium or magnesium
  • DMEM Dulbecco's minimum essential medium
  • Opti-MEM Gibco Laboratories (Life Technologies, Inc. , Grand Island, New York)
  • Bovine calf serum was obtained from Hyclone. Trypsin, EDTA (0.5 gm trypsin, 0.2 gm EDTA/liter) in Hank's salts was from Irvine Scientific.
  • NIH 3T3 cells a contact inhibited mouse fibroblast line, are from Dr. Marguerite Vogt, Salk Institute.
  • the cells are grown in monolayer in DMEM + 10% calf sera, 1% fungibact (Irvine Scientific) . Cells are removed by trypsinization with trypsin-EDTA solution. Cells are seeded onto 4 cm 2 Lab-Tek tissue culture cells per well. NIH 3T3 cells are maintained in subconfluent stock cultures to retain their contact inhibition.
  • HELA cells are a human carcinoma epithelial cell line. The cells are maintained in DMEM + 10% fetal calf sera + 1% fungibact. The cells grow by attachment to a surface and trypsinized for transfer. Cells are seeded at low density for microscope slides at approximately 1000 cells per 4 cm 2 .
  • EXAMPLE 8 Delivery of Detectable Substances into Cells Using Self-assembling Charged Lipid Complexes
  • Example 1 were diluted with water to give a concentration of 1.15 ng/ul.
  • FITC dextran, 6-carboxy fluorescein, or phalladion encapsulated in a negatively charged lipid vesicle and prepared according to Examples 4, 5 and 6 respectively, were diluted to give a concentration of approximately 0.05 ng/ul with Tris buffer (20 mM, pH 7.5; 290 mOsm) .
  • Figure 1 shows intracytoplasmic delivery of fluoresceinated dextran (MW 4000) encapsulated into negatively charged vesicles, into NIH-3T3 cells according to the methods previously described, using DOTMA/DOPE vesicles.
  • This dye is considered to be a mimic for a water soluble peptide, and its behavior would be expected to be indicative of peptide delivery.
  • Figure 2 gives similar results with HELA cells.
  • a low molecular weight fluorescent dye, 6-carboxy fluorescein, encapsulated into negatively charged vesicles can also be delivered into 3T3 cells using DOTMA/DOPE by this method as indicated on Figure 4. Again the comparison with the light micrograph illustrated co-localization with the cytoplasm of the cells.
  • Figure 5 shows the results of delivery of negatively charged vesicle encapsulated Rhodamine-phalladion, into NIH-3T3 cells using DOTAP/DOPE. Fluorescence is confined to the cytoplasm in these micrographs and the pattern of fluorescence is typical of phalladion binding to intracellular actin filaments.
  • the lipophilic dye, Rhodamine-PE, prepared as indicated in Example 6, can also be delivered intracellularly by this method, into NIH-3T3 cells using DOTMA/PE vesicles ( Figure 3) . Comparison with the light photomicrograph indicates a concentration of fluorescence intracellularly.
  • a kit was constructed to provide materials to accomplish the delivery and uptake of macromolecules into tissue culture cells.
  • the materials in the kit consisted of a vial containing dried lipid, a buffer solution, a solution containing cationic lipid vesicles, a syringe provided with a 0.4 micron NucleporeTM membrane in a filter housing, and an instruction leaflet.
  • the lipid vial contained 35 mg of l-palmitoyl-2-oleoyl-phosphatidylcholine and 15 mg of dioleoylphosphatidylglycerol in a dried lipid film.
  • the buffer solution consisted of 130 mM NaCl and 10 mM sodium phosphate buffer at a pH of 7.4.
  • the kit was used to deliver monoclonal antibodies to intercellular actin for transfection into mammalian cells according to the instructions provided.
  • a quantity of the macromolecule to be delivered in this case, 1 mg of the actin antibody, was delivered into the buffer solution and mixed well.
  • the solution was then placed in the lipid- containing vial to hydrate the lipid film and form a liposome suspension.
  • the lipid suspension was then loaded into the barrel of the syringe provided, and the liposome suspension was forced out through the filter membrane to form smaller lipid vesicles of a more uniform size. Under these conditions approximately 10% of the initial quantity of macromolecule is encapsulated into the lipid vesicles.
  • the cationic lipid vesicle solution provided in the kit consisted of 0.5 mg/ml of a DORI diether and 0.5 mg/ml dioleoylphosphatidylethanolamine.
  • the user is instructed to combine 5 ⁇ l of the negatively charged liposomes with 100 ⁇ l of the DORI/DOPE solution and to add the mixture onto tissue culture cells (approximately 10 6 cells) growing on OptiMemTM media without serum.
  • the incubation of the cells then continues for 2 hours.
  • the cells are washed, and then assayed for the biological endpoint, as in Example 8.

Abstract

Disclosed are methods and compositions for facilitating intracellular delivery of biologically active substances of pharmaceutical agents, comprising self-assembling complexes of positively and negatively charged lipid species capable of interacting with a substance to be delivered. The assembled complexes have a net positive charge suitable for spontaneously attaching to negatively charged cell membranes, and may comprise a neutral or positively charged bioactive substance first encapsulated in or complexed with negatively charged lipid vesicles which are next complexed with cationic lipid vesicles having a net positive charge before or in the process of administration to the cell.

Description

INTRACELLULAR DELIVERY OF BIOLOGICALLY ACTIVE SUBSTANCES BY MEANS OF SELF-ASSEMBLING LIPID COMPLEXES Background of the Invention The present invention relates to methods that are used to enhance delivery of "biologically and pharmacologically active agents, particularly polynucleotides, proteins, peptides, and drug molecules, by facilitating transmembrane transport or by encouraging adhesion to biological surfaces. It relates particularly to self-assembling systems, comprising lipids having cationic charged regions, that facilitate intracellular delivery of these bioactive agents.
Not all bioactive agents need to enter cells to exert their biological effect. Some are able to act on cell surfaces through cell surface receptors, while the effect of others is mediated through an interaction with extracellular components. However, many valuable therapeutic agents that are most effective in influencing cell function at the subcellular or molecular level, comprising natural biological molecules and their analogues, or foreign substances such as drugs, are preferably incorporated within the cell in order to produce their effect. In many cases, the cell membrane provides a selective barrier which is impermeable to many of these agents. The complex composition of the cell membrane comprises phospholipids, glycolipids, and cholesterol, as well as intrinsic and extrinsic proteins, and its functions are further influenced by cytoplas ic components, comprising small ions and subcellular structures. Interactions among these elements and their response to external signals make up transport processes responsible for membrane selectivity of various cells. Successful intracellular delivery of agents not naturally taken up by cells has been achieved by exploiting the natural process of membrane fusion or by exploiting the cell's natural transport mechanisms which include endocytosis and pinocytosis (Duzgunes, N. , Subcellular Biochemistry 11: 195-286 (1985)). The intracellular delivery of bioactive agents is essential for many useful applications. The delivery of polypeptides, for example, could be useful in therapies to correct genetic defects, for immunization, or in the treatment of various other disorders. Intracellular delivery of beneficial or interesting proteins can be achieved by introducing expressible DNA and mRNA into the cells of a mammal, a useful technique termed transfection. Gene sequences introduced in this way can produce the corresponding protein coded for by the gene by using endogenous protein synthetic enzymes. Therapeutic peptides that could be introduced in this way include lymphokines, such as interleukin-2, tumor necrosis factor, the interferons, growth factors, such as nerve growth factor, epidermal growth factor, and human growth hormone, tissue plasminogen activator, factor VIII:C, erythropoietin, insulin, calcitonin, and amylin. In addition, major therapeutic benefits could be obtained by means of intracellular delivery of toxic peptides such as ricin, diphtheria toxin, or cobra venom factor in order to eliminate diseased or malignant cells. Transfection of cells has been carried out by various methods, comprising calcium phosphate precipitation, or DEAE dextran or electroporation methods. Each of these methods is restrictive in some way and none is highly efficient.
Efforts to develop more satisfactory methodologies for delivering functional polynucleotides into living cells have continued steadily over the years. A major advance was the development of cationic lipid technology, based on a discovery that a positively charged synthetic cationic lipid, N- [ 1- ( 2 # 3-dioleyloxy)propyl]-N,N,N- trimethylammonium chloride (DOTMA) , in the form of liposomes, or small vesicles, could interact spontaneously with DNA, which is negatively charged, or anionic, to form lipid-DNA complexes having a net positive charge and capable of fusing with the negatively charged cell membranes of tissue culture cells, to achieve both uptake and expression of the DNA (Feigner, P.L. et al. Proc. Natl. Acad. Sci.. USA 84:7413-7417 (1987) and U.S. Patent No. 4,897,355 to Eppstein, D. et al.). The Lipofectinw reagent (Bethesda Research Laboratories, Gaithersburg, Maryland) , an effective commercial cationic lipid reagent for the delivery of highly anionic polynucleotides into living tissue culture cells, comprises positively charged DOTMA liposomes.
Since the introduction of DOTMA, other cationic lipid agents have been developed for use in transfeeting cells. A
DOTMA analogue, 1,2-bis(oleoyloxy)-3-(trimethylammonio)- propane (DOTAP) has been successfully used in combination with a phospholipid to form DNA-complexing vesicles
(Stamatatos, L. et al.. Biochemistry 27(11) :3917- 3925(1988). Also a co-pending U.S. Patent Application
Cationic Lipids for Intracellular Delivery of Biologically
Active Molecules, filed April 19th, 1990 by Feigner et al., discloses cationic lipids which are more effective and less toxic than presently known cationic lipids for transfection. Cationic lipid methodology is presently preferred over other methods; it is more convenient and efficient than calcium phosphate, DEAE dextran or electroporation methods. However, it can only be applied to polyanionic molecules, such as DNA. Unfortunately, not all bioactive substances of interest are as negatively charged as polynucleotides; therefore, the extension of cationic lipid methodology to the delivery of other molecules, for example, many proteins, is not feasible.
Transport of proteins is among the most selective and specific of transmembrane processes. Such processes involve protein-protein recognition (signal sequences) , frequently require ATP, or are enzymatically driven. Viruses have proteins capable of accessing these processes successfully, thus allowing viral infections of cells to occur. The viral method of intracellular penetration has been exploited to develop a methodology which uses viral "chimerasomes" or proteoliposomes for intracellular protein delivery. Viral chimerasomes are lipid vesicles containing molecules intended for intracellular delivery together with two critical proteins isolated from viruses. One of these proteins is a cell surface recognition protein which promotes attachment of lipid vesicles with the cell surface. Another is a viral fusion protein which facilitates the introduction of the viral contents into the cell cytoplasm. Unlike the Lipofectin1" reagent, the chimerasome methodology is broadly applicable to the delivery of proteins as well as polynucleotides. However, it is more expensive and difficult to manufacture than cationic lipid reagents because it involves isolation and purification of components of potentially infectious virus particles. Stability of the proteins and lipids in the chi erosome reagent is a potential problem, and it is also somewhat less convenient to use than the cationic lipids.
A modification of the cationic lipid technology has been suggested which may confer a degree of specificity on its transfection procedures similar to that of chi erosomes. According to the strategy of Eppstein, D. et al. U.S. Patent No. 4,897,355, polynucleotide molecules are first treated with DOTMA to produce a positively charged complex. This complex is then treated with negatively charged liposomes to form a double coated, negatively charged complex. The negatively charged coat may comprise a coupling reagent. Receptor specific agents, such as antibodies, lectins, and other ligands may then be attached to the negatively charged outer layer through the coupling reagent. The overall negative charge of the double-coated complex prohibits its non-specific interaction with the negatively charged membranes of cells while allowing relatively specific interaction through the binding of integral ligands. The defect of this approach, like that of the chimerosome methodology, is its relative complexity, and its requirement for specific binding reagents; further, its application, like that of the parent cationic lipid methodology, is limited to polyanionic materials, that is. polynucleotides and negatively charged proteins.
It would be desirable to have an intracellular delivery system having the advantages of the above described cationic lipid and chimerosome methodologies but without their inherent limitations.
It is therefore an object of the invention to provide a methodology for the intracellular delivery of a wide variety of bioactive agents, particularly proteins, regardless of net charge. The intracellular delivery systems disclosed herein exploit self-assembling processes found in natural systems to join together molecular subcomplexes through physical mechanisms of ionic attraction and repulsion and hydrophobic-hydrophilic interactions. The most pertinent example of such systems in nature is the simple assemblies of lipids and proteins to form cell membranes. The physical forces holding such structures together are relatively weak and non-specific, and this, among other factors, allows the assembly to proceed spontaneously and rapidly.
An advantage of self-assembling systems that imitate these natural systems is that the energy conserving nature of their assembly and disassembly eliminates the necessity of chemically bonding the subunits together or providing means to degrade them to the active units in vivo. The property of self-assembly therefore enables a convenient and practical manufacturing procedure.
It is also an object of the invention to provide a generally applicable intracellular delivery system which is self-assembling, eliminating the need for synthesis procedures.
It is further an object of the invention to provide reliable and efficient reagents for an intracellular delivery system which are commercially feasible, that is, relatively easy and inexpensive to manufacture and having an extended shelf life. Brief Description of the Drawings
The following figures show the intracellular delivery of proteins into tissue cells in culture according to the methods of the invention. Each figure consists of a pair of photomicrographs produced by (a) phase contrast and (b) fluorescence microscopy.
Figure 1 shows intracytoplasmic delivery of fluoresceinated dextran (MW 4000) , into NIH-3T3 cells. Figure 2 shows intracytoplasmic delivery of fluoresceinated dextran (MW 4000) into HELA cells.
Figure 3 shows the intracellular delivery of Rhoda ine-PE into NIH-3T3 cells.
Figure 4 shows the delivery of a low molecular weight dye, 6CF, into NIH-3T3 cells.
Figure 5 shows the delivery of Rhodamine-phalloidin into NIH-3T3 cells.
Summary of the Invention
The invention provides a method for making a vehicle for administering a bioactive substance to a cell, comprising the steps of providing the substance in a first lipid vesicle which vesicle comprise at least one negatively charged lipid and is negatively charged; and combining this first lipid vesicle with second lipid vesicles comprising at least one cationic lipid and having a net positive charge, whereby the first negatively charged vesicles are coated with at least one positively charged lipid vesicle. The bioactive substance can be encapsulated in the vesicle by containment in the aqueous core; alternatively, if the bioactive substance is lipophilic or has a lipophilic or amphipathic region, it may be incorporated by means of that lipophilic structure into the lipid wall of the vesicle.
In another embodiment, there is provided a method for making a vehicle for administering a bioactive substance to a cell, comprising the steps of forming a first complex comprising a positively charged macromolecule and at least one lipid vesicle, wherein the lipid vesicle comprise a negatively charged lipid and has a net negative charge; and contacting the first complex with lipid vesicles comprising at least one cationic lipid and having a net positive charge to form a second complex.
According to another aspect of the invention, there is provided a method for administering a bioactive substance to a cell, comprising the steps of providing the substance in a negatively charged lipid vesicle; and administering the negatively charged lipid vesicle to a cell together with a lipid vesicle comprising at least one cationic lipid and having a net positive charge.
In another embodiment there is provided a method for administering a bioactive substance to a cell, comprising the steps of preparing a complex comprising said bioactive substance and a negatively charged lipid vesicle; and administering the complex to a cell together with a lipid vesicle comprising at least one cationic lipid and having a net positive charge. The methods of administering bioactive substances to a cell can be applied in vivo as well as in vi tro .
The invention also provides a composition of matter, comprising a bioactive substance contained in a negatively charged lipid vesicle and lipid vesicles, comprising at least one cationic lipid species and having a net positive charge, at least partially covering the negatively charged lipid vesicle. In preferred embodiments, the composition has a net positive charge. The bioactive substance of the composition can be a biological macromolecule, such as, for example, a polynucleotide, a protein or polypeptide. The bioactive substance may also be a drug, and the drug may be a small organic molecule. According to one preferred embodiment, a bioactive substance is encapsulated within a negatively charged lipid vesicle. The bioactive substance may also be lipophilic or comprise a lipophilic region, and is thereby incorporated among the lipids of the lipid vesicle.
The invention also provides a kit, comprising materials, supplies, reagents, and instructions for preparing self-assembling lipid complexes for the delivery of a bioactive substance to a cell, either in vi tro or in vivo , comprising at least one vessel containing at least one negatively charged amphipathic lipid species; and a quantity of a lipid substance, the substance comprising at least one positively charged amphipathic lipid species. The kit can further comprise at least one hydrating buffer solution and an instruction leaflet.
In any of the methods or compositions disclosed suitable positively charged lipid species can be known or unknown cationic lipids, such as, for example DOTAP orDOTMA or can have the structure of any of the novel cationic lipids described herein.
Detailed Description of the Invention
The present invention provides vehicles, comprising self-assembling lipid complexes, capable of delivering bioactive substances, particularly polynucleotides, oligonucleotides, proteins, peptides or drugs into living cells in a convenient and efficient manner. According to the methods presented, lipid vesicles, comprising lipid species having either a negative or positive charge, are combined with the substance to be delivered in a properly ordered sequence, forming self-assembling lipid complexes containing the substance to be delivered and having a positively charged outer lipid layer.
The invention, like cationic lipid technology, takes advantage of the natural fusion properties of a positively charged lipid to promote passage of bioactive substances associated with that lipid across the negatively charged cell membrane. However, the invention provides a prior association of the bioactive substance with negatively charged lipids to form primary complexes having a net negative charge. This primary complex promotes a subsequent secondary complex formation with the liposomes comprising the cationic lipids that facilitate membrane transport. In this way the invention extends cationic lipid methodology to all bioactive agents intended for intracellular delivery, regardless of size or net charge. It also provides an intracellular delivery process that is quite non-specific and is therefore broadly applicable to all cell types, in cases where these substances, as such, are not able to penetrate the cell membrane, including those of cells that do not contain specific ligand or antibody receptors. Lipid Vehicles for Intracellular Delivery
Vehicles for intracellular delivery according to the invention comprise a primary negatively charged lipid complex in association with the bioactive substance to be delivered and a secondary complex, having a net positive charge and comprising the primary negative complex in association with positively charged liposomes. The invention provides two general strategies for assembling the primary negatively charged lipid complex comprising the bioactive substance. According to a preferred strategy, the bioactive substance to be delivered is entrapped within a lipid vesicle or liposome comprising negatively charged lipids and having a net negative charge. The entrapment can occur, according to one embodiment of the invention, as an incorporation of a lipophilic bioactive substance, or such a substance having a lipophilic region, into the wall of the lipid vesicle. According to another embodiment, the entrapment can occur as an encapsulation of hydrophilic bioactive substance into the aqueous core of the lipid vesicle. For both lipophilic and hydrophilic substances, the uptake into the negatively charged liposome occurs during the process of liposome formation, typically the hydration of a lipid film, disclosed herein, and illustrated by examples. Examples 3, 5, and 6 disclose the encapsulation of substances within negatively charged liposomes, and Example 4 discloses the incorporation of a lipid derivative into the liposome wall.
According to another strategy, the bioactive substance is contacted with negatively charged liposomes, and these liposomes then at least partially enclose the substance to provide a primary lipid complex having negatively charged regions. In a preferred embodiment, the bioactive substance has a net positive charge and interacts with the negative liposomes by means of charge attraction; alternatively, the bioactive substance can be neutral or negatively charged but comprise lipophilic regions that promote its association with the liposomes of the complex through hydrophobic or lipophilic interactions. In yet another embodiment, the bioactive substance can comprise amphipathic peptide helices, for example of the type disclosed by Segrist, U.S. Patent No. 4,643,988, that can attach to the lipid vesicle surface through hydrophobic and lipophilic interactions, thus anchoring the molecules of which they are a region. In other preferred embodiments, the negatively charged liposomes used to form the complex may contain the same or other bioactive substances, either encapsulated or incorporated, as disclosed above.
The second element of the intracellular delivery system of the invention comprises liposomes formed from positively charged cationic lipids and having a net positive charge. On contact with the primary complex, comprising either the negatively charged liposomes or the negatively charged lipid complex, the positively charged liposomes spontaneously form complexes having a net positive surface charge.
In all the embodiments described above, the positively charged assembled complexes, when placed in contact with cells, will spontaneously attach to cell surfaces and interact directly with the plasma membrane, according to conventional cationic lipid methodology. Delivery of the bioactive cargo into the cell cytoplasm can occur by various mechanisms. For example, the lipids of the complex may either directly fuse with the plasma membrane and discharge the entrapped substance intracellularly; alternatively, it may be phagocytized and interact with other internal membranes or the membranes of the phagocytic compartment itself.
All of these assemblies, comprising the encapsulation or incorporation of substances within lipid vesicles, the association of substances with positively or negatively charged lipids or vesicles thereof, the association of positively and negatively charged lipids and vesicles thereof, and even the attachment of the vesicles to the target cell surface and fusion directly with cell membranes, occur spontaneously as the result of repulsions and attractions of ionic charges and hydrophobic-hydrophilic interactions.
Therefore, the negative and positively charged lipid complexes form almost instantaneously, and accordingly the primary negatively charged complexes can be contacted with the positively charged liposomal preparation and target cells simultaneously. Alternatively, the primary negative complexes and positively charged liposomes may be mixed to form secondary complexes before contact with the target cells. For in vivo applications, prior complex formation is preferred, while for in vi tro applications, the positively charged empty liposomes are conveniently added to a cell culture at the same time as the negatively charged liposome-bioactive agent complexes, thus avoiding a separate mixing step. Liposome Formulations
The lipid reagents of the invention may comprise lipid mixtures similar to that of the physiological cell membrane, comprising phospholipids as primary components. The lipid reagents can further comprise any of the conventional synthetic or natural liposome materials, including phospholipids from natural plant or animal sources such as phosphatidylcholine . phosphatidylethanolamine, sphingomyelin, phosphatidylserine, or phosphoinositol. Synthetic phospholipids that may also be used include, but are not limited to, dimyristoylphosphatidylchol ine , dioleoylphosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphatidylcholine and the corresponding synthetic pho s ph at i dy 1 e th ano 1 ami ne s and phosphatidylglycerols. Other additives such as cholesterol, glycolipids, fatty acids, sphingolipids, or gangliosides can also be used, as is conventionally known for the preparation of liposomes.
The positively and negatively charged lipid vesicles used in the methods of the invention are typically prepared as appropriate from a mixture of either cationic lipids or negatively charged lipids, neutral lipids and cholesterol or a similar sterol. Neutral lipids can be phosphatidylcholine, phosphatidyl ethanolamine, similar phospholipid analogs, or mixtures of these, as well as monoglycerides, diglycerides and triglyσerides. The negatively charged lipid reagents of the invention are those comprising at least one lipid species having a net negative charge at physiological pH or combinations of these. Suitable lipid species comprise phosphatidyl glycerol and phosphatidic acid or a similar phospholipid analog. The positively charged lipid reagents of the invention are those comprising at least one cationic lipid species having a net positive charge at physiological pH. Suitable lipid species comprise known cationic lipids, such as l,2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP) or N-(ω, ω-l-dialkoxy)-alk-l-yl-N, N, N-trisubstituted ammonium surfactants, such as DOTMA, or complex cationic lipids having similar structures and properties or mixtures of these. Particularly preferred cationic lipids are those disclosed in a co-pending U.S. Application entitled Cationic Lipids for Intracellular Delivery of Biologically Active Molecules filed April 19th, 1990 by Feigner et al., which is hereby incorporated by reference. These cationic
Figure imgf000015_0002
Figure imgf000015_0001
wherein
Y1 and Y- are the same or different and are -0-CH2- -
O-C(O)-, or -0-; R1 and R2 are the same or different and are H, or C-_ to C23 alkyl or alkenyl;
R3 and R4 are the same or different and are Ci to C24 alkyl, or H;
R5 is Ci to C24 alkyl straight chain or branched chain; R6 is -C(0)-(CH2)m_NH-, a diaminocarboxylic acid which is alkyl, aryl, or aralkyl, or -C(O)-(CH2)m~NH~ linked to said diaminocarboxylic acid, or is absent;
R7 is H, spermine, spermidine, a histone, or a protein with
DNA-binding specificity, or wherein the amines of the R7 moiety are quaternized with R3, R4, or R5 groups; or
R7 is an L- or D-alpha amino acids having a positively charged group on the side chain, said amino acids comprising arginine, histidine, lysine or ornithine or derivatives thereof, or the same amino acids wherein the amine of the R7 moiety is quaternized with R3, R4 or R5 groups; or
R7 is a polypeptide selected from the group consisting of
L- or D-alpha amino acids, wherein at least one of the amino acids residues comprises arginine, histidine, lysine, ornithine, or derivatives thereof; n is 1 to 8; m is 1 to 18; and
X is a non-toxic anion.
These compounds have been found to be highly effective for use in lipid formulations for transfection and other intracellular delivery procedures. Particularly preferred for in vivo transfection or other cell delivery are the diester or ether/ester species of these compounds which are found to be more easily metabolized than previously known cationic lipids while nonetheless retaining a high level of tranεfective activity. In particularly preferred embodiments, therefore, the positively charged liposomes of the invention comprise the cationic lipids l,2-dioleoyl-3- dimethylaminopropyl-j8-hydroxyethylammonium or 1-0-oleyl-, 2-oleoyl-3-dimethylaminopropyl-0-hydroxyethylammonium.
For the same reason of metabolizability, ester or ester derivatives of the known cationic lipids having a formula as set forth below are also preferred.
Figure imgf000016_0001
or an optical isomer thereof, wherein
Y1 and Y2 are different and are either -0-CH2-, -O-C(O)-, or OH;
R1 and R2 are individually absent or are Ci to C23 alkyl or alkenyl; R3, R4 and R5 are the same or different and are H, C^ to
C14 alkyl, C7 to Cn aryl or alkaryl, or at least two of
R3, R4, and R5 are taken together to form quinuclidino, piperidino, pyrrolidino, or morpholino; n is 1 to 22; and X is a non-toxic anion.
Non-toxic anions described herein may be those of pharmaceutically non-toxic acids including inorganic acids and organic acids. Such acids include hydrochloric, hydrobromic, sulfuric, phosphoric, acetic, benzoic, citric, glutamic, lactic acid and the like. For the preparation of pharmaceutically acceptable salts, see S. M. Berge et al.. Journal of Pharmaceutical Sciences. 66:1-19(1977) which is incorporated herein by reference.
In a lipid formulation for preparing positively charged lipid vesicles, the cationic lipid can be present at a concentration of between about 0.1 mole% and 100 mole%, preferably 5 to 95 mole%, and most preferably between 20 and 80 mole%. In a formulation for preparing negatively charged lipid vesicles, the negatively charged lipid can be present at a concentration between about 0.1 and 100 mole%, preferably 1 to 90 mole%, and most preferably 3 to 50 mole%.
In order to produce lipid vesicles having a net charge, the quantity of the positively or negatively charged component must exceed that of the alternatively charged component. The alternatively charged lipid can be present at between about 0 to 49 mole% and preferably 0 to 40 mole%.
The neutral lipid can be present in positively or negatively charged lipid vesicles in a concentration of between about 0 and 99.9 mole %, preferably 5 to 95 mole%, and most preferably 20 to 80 mole%. Cholesterol or a similar sterol can be present at 0 to 80 mole %, and preferably 0 to 50 mole%. The lipid reagents may be prepared and stored as empty liposomes in aqueous solution, or may be stored as dried lipid, for example as a lipid film, after formulation to be later used as encapsulating reagents for selected bioactive substances. Liposome Formation
It should be understood that the lipid formulations of the invention, comprising at least one amphipathic lipid, such as a phospholipid, spontaneously assemble to form primary liposomes, heterogeneous in size and structure, in aqueous solution. Therefore the term lipid reagent, lipid vesicles, and liposomes are used interchangeably in describing formulations. The lipid reagents, either negatively or positively charged, are prepared as liposomes or lipid vesicles, according to Example 1. The component lipids are dissolved in a solvent such as chloroform and the mixture evaporated to dryness as a film on the inner surface of a glass vessel. On suspension in an aqueous solvent, the amphipathic lipid molecules assemble themselves into primary liposomes. If other molecules are present in the aqueous solvent, such as, for example, a bioactive substance, these will be captured within the liposomes, as demonstrated in Examples 3 and 5. Alternatively, some lipophilic bioactive substances intended for entrapment can be dissolved or suspended in the organic solvent that is used to dissolve the lipids of the liposome formulation. After evaporation of the lipid film and hydration to form primary liposomes, the substances thus dissolved are typically incorporated into the bilayer of those liposomes, rather than being encapsulated into the aqueous interior. Otherwise, if there is no solute in the hydration buffer, empty liposomes will be formed, as demonstrated in Examples 1 and 2.
To prepare liposomes suitable for physiological in vivo use, having a unilamellar structure and a uniform size of from about 50 to about 200 μm in diameter, the primary liposomes are preferably processed by the freeze-thaw and extrusion processes, as provided by Example 2. These primary liposomes are reduced to a selected mean diameter by means of the freeze-thaw procedure referred to above. The cationic lipids of the invention are formed into vesicles of uniform size prior to transfection procedures, according to methods for vesicle production published in the literature and knov/n to those in the art, for example, the sonication of spontaneously formed liposomes comprised of the lipids in aqueous solution described by Feigner, P.L. et al., Proc. Natl. Acad. Sci.. USA 84:7413-7417 (1987) or the reverse-phase evaporation procedure of J. Wilschut et al. Biochemistry 19:6011-6021(1980) or freeze- thaw and extrusion (Mayer, L. et al., Biochim. Biophys. Acta 858:161-168 (1986).
Suitable conventional methods of preparation include, but are not limited to, those disclosed by Bangham, A. et al., J. Mol. Biol. 23: 238-252 (1965); Olson, F. et al., Biochim. Biophys. Acta 557: 9-23 (1979), Szoka F. et al., Proc. Natl. Acad. Sci. USA 75: 4194-4198 (1978), Mayhew, E. et al. Biochim. Biophys. Acta 775: 169-175 (1984), Kim, S. et al. Biochim. Biophys. Acta 728:339-348) , and Mayer, L. et al., Biochim. Biophys. Acta 858:161-168 (1986).
According to a preferred method, the negative and positive lipid reagents of €he invention, comprising at least one amphipathic lipid species, such as a phospholipid, are prepared using the freeze-thaw-extrusion procedure indicated in Examples 1 and 2. The component lipids are dissolved in a solvent such as chloroform and the mixture evaporated to dryness as a film on the inner surface of a glass vessel. On suspension in an aqueous solvent, the amphipathic lipid molecules assemble themselves into primary liposomes. If other molecules are present in the aqueous solvent, such as, for example, a bioactive substance, these will be captured within the liposomes, as indicated in Examples 3, 4, 5 and 6. Otherwise, empty liposomes will be formed, as in Example 1. These primary liposomes are reduced to a selected mean diameter by means of the freeze-thaw procedure described in Example 2.
The bioactive cargo of the lipid delivery system, according to the primary strategy of the invention, is entrapped into negatively charged liposomes. The lipid derivatives of agents disclosed above, for example, the lipid derivatives of antiviral nucleosideε such as phosphatidylazidc nymidine or (3'-azido-3,-deoxy)thymidine- 5'-diphospho-3-diacylglycerol, can be entrapped by direct incorporation into the wall of the lipid vesicle during the hydration of a lipid film comprising these lipid derivatives in the liposome formulation. Other agents can be encapsulated within the aqueous space of the liposome according to conventional liposome forming methodology, as follows.
Encapsulation Procedure Substances intended for intracellular delivery can be encapsulated into negatively or positively charged liposomes by any one of a number of standard procedures. For example, each of the lipid components comprising the liposomal formulation is dissolved together into a co- miscible organic solvent. If a lipid derivative of an agent to be delivered is to be incorporated into the liposome, a selected quantity of that agent is added to the other lipid components at this time. The solvent is evaporated and the vessel containing the residual lipid film is evacuated overnight to remove solvent traces.
An hydration solution is next added to the dried lipid film to form primary liposomes. If hydration buffer alone is added to a film of lipid components alone, empty liposomes will form. If hydration buffer alone is added to a lipid film comprising lipid derivatives of bioactive agent, the primary liposomes will comprise that agent incorporated into the vesicle walls. If hydration buffer having a bioactive dissolved therein is added to a lipid film comprising only lipids, the primary liposomes that form will comprise that agent trapped in the interior aqueous compartment of the liposomes. Permutations of the processes, resulting in liposomes comprising bioactive agents both incorporated into the walls of the vesicle as well as entrapped therein are possible and are within the contemplation of the invention.
The hydration solution can be any biologically compatible buffer solution comprising isotonic saline or phosphate buffered saline, or low ionic strength buffers comprising 5% sorbitol or 10% sucrose. Such buffers are well known to those skilled in the art. The concentration of the bioactive substance in the hydration buffer which is intended for intracellular delivery can vary widely depending on the substance or the application; this concentration can be between 1 picogram/ml and 500 mg/ml. Following hydration of the lipid film, the resulting liposome suspension can be further emulsified by any one of a number of procedures; for example, the sample can be forced through Nuclepore'" membranes to produce vesicles of a size comparable to the pore size of the membranes. Encapsulations are described in Examples 3, 4, and 6 for the encapsulation of FITC dextran, 6-carboxy-fluorescein, and phalladion in the negatively charges lipid formulation of 4.9/2.1/3 DOPC/DOPG/cholesterol, followed by freeze- thaw and extrusion to achieve a uniform sized liposomal preparation. Any of the substance remaining unencapsulated can be removed, if desired, by a process such as gel filtration chromatography.
The procedure for delivering materials into cells according to the method of the invention comprises a strategy of presenting the material to the cell in association with positively and negatively charged lipid vesicles, described as follows, and illustrated by the accompanying Examples.
Intracellular Delivery of Liposome Associated Bioactive Substances
In a preferred embodiment of the method of the invention, following the entrapment of a bioactive substance into negatively charged liposomes, or association of the substance with empty negatively charged liposomes, the resulting liposomes or complexes are added directly to cells, for example in an in vi tro application, in a suitable biologically compatible medium, together with positively charged lipid vesicles. The concentration of lipid with which the cells are contacted varies widely depending on the application, but is between 1 μ olar and
50 mmolar and preferably from 10 μmolar to 10 mmolar. The positively charged lipid vesicles attach spontaneously to the target cell surface, fuse with the cellular membranes and deliver the contents of the liposome into the cell cytopiasm.
In an alternate approach, the substance intended for intracellular delivery is first encapsulated into negatively charged liposomes, or complexed with empty negatively charged liposomes, according to the procedures described previously. These primary negative complexes are then exposed to a quantity of positively charged lipid vesicles. Immediately upon mixing the two solutions containing the oppositely charged lipid vesicles, complexes spontaneously form comprising negatively and positively charged vesicles.
The quantity of positively charged vesicles added to the encapsulating negatively charged vesicles should be sufficient to encourage attachment of the complexes to the target cell surface. Theoretically, this quantity should be sufficient to provide the complexes with a net positive charge, with the number of positive charges contributed by the positively charged liposomes in excess over the number of negative charges contributed by the negatively charged liposomes; however, such an excess of positively charged elements is not always required to encourage sufficient attachment to the target cell surface to achieve intracellular delivery. Accordingly, the ratio of positive to negative charges in the final complexes may be from about 100:1 to 0.1:1 and preferably 20:1 to 0.2:1.
Intracellular Delivery of Liposome-Associated Polynucleotides
Procedures for the encapsulation and intracellular delivery of polynucleotide using cationic lipid methodology frequently result in a high percentage of the polynucleotide remaining unencapsulated by lipid vesicles. The present invention also contemplates the use of liposome complexes comprising positively charged liposomes but having a net overall negative charge for the intracellular delivery of negatively charged polynucleotides. These complexes are prepared as follows: Polynucleotides in solution are mixed with a sufficient quantity of positively char:sd liposomes so as to form a complex which reduces the anionic characteristics of the polynucleotide by a sufficient amount as described below. Negatively charged elements in a polynucleotide solution can be readily quantified using the extinction coefficient for the nucleotide monomer which bears one negative charge per monomer. The concentration of polynucleotide in solution can be from about 0.01 μg/ml to 50 mg/ml, preferably from 1 μg/ml to 10 mg/ml and most preferably from 10 μg/ml to 1 mg/ml. The concentration of positively charged lipid vesicles can range from between 0.1 μg/ml to 100 mg/ml, preferably from 1 μg/ml to 100 mg/ml, and most preferably from 10 μg/ml to 50 mg/ml. The solutions may be mixed together from those having a low ionic strength buffer, that is having an ionic strength less than that of 25 mM sodium chloride. Sorbitol, sucrose or glucose can be used to render a low ionic strength buffer isotonic. Adsorption of the polyanionic polynucleotides to the cationic vesicles reduces the negative charge character of the polynucleotides. In theory, the quantity of charges contributed by the vesicles should exceed the number of negative charges contributed by the polynucleotide, although this condition may not be an absolute requirement for every application. The ratio of positive to negative charges in the polynucleotide/cationic lipid complexes may be from about 100:1 to 0.1:1 and preferably 20:1 to 0.2:1.
These positively charged complexes can further spontaneously associate with an excess of negatively charged vesicles so as to produce a negatively charged complex containing positively charged lipids and the polynucleotide. By following a protocol of this type, the entire polynucleotide can be incorporated by self-assembly into a lipid complex that can accomplish intracellular delivery without a loss in efficiency caused by incomplete nucleotide encapsulation. In theory, the quantity of negatively charged liposomes added should exceed the net positive charge contributed by the complex of positively charged lipid and polynucleotide, although in practice this level of negatively charged elements may not be required for every application. Accordingly, the ratio of positive to negative charges in the polynucleotide/cationic lipid complexes may be 100:1 to 0.1:1 and more preferably 20:1 to 0.2:1.
The negatively charged complexes produced above, containing polynucleotide and positively charged lipids, are analogous to the primary negatively charged lipid complexes disclosed for other bioactive substances, and can be delivered in vivo or in vi tro to tissue culture cells in a manner similar to that described above for those primary complexes.
In a parallel manner, self-assembled systems comprising successive layers of positive and negative liposomes can be manufactured and can be applied to any molecule or composition to be delivered to a cell, regardless of its size or charge. The construction of such structured complexes would be otherwise be extremely difficult.
In the Examples that follow, intracellular delivery of proteins was measured by using signal producing substances that could be visualized in the cells after incorporation. These substances include:
(1) Fluoresceinated dextran which is available in different molecular sizes from 4,000 to 70,000 MW and having a net positive, neutral or negative charge. Delivery of dextran into the cell was quantitated by total cell associated fluorescence as determined by a spectrophotometer. The distribution of fluorescence was determined by fluorescence microscopy. A thin band of fluorescence around the cell was read to indicate cell-associated vesicles; bright spots around the nucleus to indicate lysosomal uptake; and diffuse cytoplasmic fluorescence to indicate cytoplasmic delivery. Cytoplasmic delivery is considered the most effective.
(2) Fluoresceinated phalloidin. This 7-amino acid cyclic peptide binds specifically to intracellular actin. Functional delivery of this substance is indicated by the filamentous intracellular staining pattern characteristic of actin filaments seen on examination of the cells by fluorescence microscopy.
(3) Anti-actin antibody. This antibody will bind to actin when delivered intracellularly. Treated cells were fixed, permeabilized and counter-stained with a fluorescent antibody against the anti-actin antibody. As in the case of fluoresceinated phalloidin, effective delivery is indicated by the characteristic actin pattern seen on fluorescence microscopy. Cell viability for all experiments was determined by trypan blue staining.
The results indicate that a variety of biologically significant materials can be delivered by this procedure. Utility
The self-assembling delivery systems of the invention can be advantageously formed using any cationic lipid, whether those previously known, for example, DOTMA or DOTAP, or novel cationic lipids, such as those described above and also disclosed in co-pending U.S. Application of Feigner et al., filed April 19, 1990, to deliver substances intracellularly either in vitro or in vivo. Those cationic lipids described above having metabolizable ester bonds are preferred for in vi vo use. In vi tro applications include the intracellular delivery to any cell grown in culture, comprising cells of any species, whether plant or animal, vertebrate or invertebrate, and of any tissue or type.
Contemplated uses comprise transfection procedures corresponding to those presently known. Accordingly, the strategies of the invention can be used to facilitate the intracellular delivery of DNA or mRNA sequences coding for therapeutically active polypeptides, as described in detail in U.S. Patent Applications Serial Nos. 326,305 and 467,881 which are hereby incorporated by reference. The self- assembling delivery methods described herein however, are particularly preferred for the delivery of the expressed gene product or protein itself. Thus the self-assembling delivery of proteins can provide therapy for genetic disease by supplying deficient or absent gene products to treat any genetic disease in which the defective gene or its product has been identified, such as Duchenne's dystrophy (Kunkel, L. and Hoffman, E. Brit. Med. Bull. 45(3) :630-643 (1989) or cystic fibrosis (Goodfellow, P. Nature. 341(6238) :102-3 (Sept. 14, 1989).
The self-assembling delivery systems described above can also provide immunizing polypeptides to the cell either by delivering a polynucleotide coding for the immunogen, or the immunogen itself. Other therapeutically important polynucleotides suitable for self-assembling delivery comprise anti-sense polynucleotide sequences, useful in eliminating or reducing the production of a gene product, as described by Ts'o, P. et al. Annals New York Acad. Sci. 570:220-241 (1987). Also within the scope of the invention is the delivery, by means of the methods disclosed, ribozymes, or catalytic RNA species, either of the "hairpin" type as described by Ha pel et al. Nucleic Acids Research 18(2) :299-304 (1990); or the "hammerhead" type described by Cech, T. and Bass, B. Annual Rev. Biochem. 55:599-629 (1986) .
Particularly preferred within the contemplated uses of the invention are deliveries of either an anti-sense polynucleotide or ribozy e as described above, and having as its target the rev site if the HIV genome (Scientific American, October, 1988, pp. 56-57). Matsukura, M. et al. Proc. Natl. Acad. Sci. 86:4244-4248 (1989) describe a 28-mer phosphorothioate compound anti-HIV (anti-rev transactivator) specific for the site. The transfection procedures described above may be applied by direct injection of self-assembled lipid complexes, comprising DNA, RNA or proteins into cells of an animal in vi vo . However, these methods are particularly effective at facilitating in vi tro transfection of cells. Therefore the above therapies can be alternatively carried out by in vi tro transfection of some of the cells of an animal using self-assembling delivery methods, and re- introduction of the cells into the animal. The ability to transfect cells at high efficiency with self-assembling delivery thus provides an alternate method for immunization. The gene for an antigen is introduced into cells which have been removed from an animal by self- assembling delivery methods. The trans-fected cells, now expressing the antigen, are reinjected into the animal where the immune system can now respond to the (now) endogenous antigen. The process can possibly be enhanced by co-injection of either an adjuvant or lymphokines to further stimulate the lymphoid cells.
The compositions and methods of the present invention can be most advantageously used in the periodic supplying of endogenous or exogenous macromolecules, particularly proteins, to a cell. They are therefore particularly suitable for use in transient therapies which requires treatment with proteins, particularly in cells unable to carry out translation of messenger RNA.
Examples of such transient therapeutic uses of self- assembling delivery methods herein disclosed include the liposomal delivery of nucleotide analogues having an antiviral effect, such as dideoxynucleosides, didehydronucleosides, nucleoside analogues having halogen substituted and azido-substituted ribose moieties, such as 3**-azido-3,deoxythymidine (AZT) , or nucleoside analogues such as acyclovir or gancyclovir (DHPG) . Lipid derivatives of antiviral nucleosides are presented in U.S. Patent
Applications Serial Nos. 216,412; 319,485; and 373,088 which are hereby incorporated by reference. These lipid derivatives are conveniently incorporated into the vesicle walls of the primary negatively charged liposomes according to the primary strategy of the invention. Among other therapeutically important agents that can be thus delivered are peptides comprising physiologic species such as interleukin-2, tumor necrosis factor, tissue plasminogen activator, factor VIII:C, erythropoietin, growth factors such aε epidermal growth factor, neural growth factor, and hormones such as tissue insulin, calcitonin, and human growth hormone as well as toxic peptides such as ricin, diphtheria toxin, or cobra venom factor, capable of eliminating diseased or malignant cells. Also within the scope of the invention is the intracellular delivery of antiviral formulation through topical application of the lipid complexes disclosed comprising acyclovir or gancyclovir for the treatment of Herpes simplex. These formulations preferably comprise lipid derivatives of the antiviral agents, particularly the phosphatidylglycerol derivatives as disclosed in U.S. Applications Serial Nos. 216,412, 319,485 and 373,088. The effect of the self-assembled lipid delivery system is to facilitate the penetration of the active antiviral agent through the stratum corneum of the dermis.
Self-assembling systems comprising lipid-encapsulated bioactive agents may accordingly be used for intracellular delivery either in vi vo or in vi tro . For in vi tro applications, neutral or positively charged bioactive molecules are encapsulated in a negatively charged lipid reagent and added to a washed cell culture together with a volume of a suspension of liposomes comprising the positively charged lipid reagent, as indicated in Example 8. For in vivo applications, corresponding molecules are encapsulated with negatively charged lipid reagent and then coated with the liposomes of the positively charged lipid reagent before administration.
EXAMPLE 1: Positively Charged Liposome Preparation
Lipids used: l,2-Bis(oleoyloxy)-3-(trimethylammonio) propane, DOTAP; dioleoyl phosphatidylethanolamine, DOPE.
A quantity of 5 mg of DOTAP was combined with 5 mg DOPE in 1 ml chloroform, dried in a Rotovap and the flask evacuated overnight on a vacuum pump. The lipids were then suspended in 0.5 ml double deionized water, and sonicated in a bath sonicator for 30 minutes at 15° or until the suspension was clear. A clear solution is indicative of liposome formation. (When the lipid concentration is greater than 80 mg/ml the solution does not clear.)
EXAMPLE 2: Empty Negatively Charged Liposomes
Lipids used: dioleoylphosphatidylcholine, DOPC; dioleoylphosphatidylglycerol, DOPG; cholesterol Quantities of 4.67 mg DOPC, 4.06 mg DOPG and 2.9 mg cholesterol comprising 25 uMoles of total lipid, in a molar ratio of 4.9/2.1/3, DOPC/DOPG/cholesterol, were combined in 1 ml of chloroform. The mixture was dried on a Rotavap apparatus and the flask evacuated overnight on a vacuum pump.
The lipid film was suspended at 20"C for 1 hour using a rotary shaker with l ml tris buffer (20 mM pH 7.5) and the suspension adjusted to 290 mOsm by adding crystalline NaCl. The sample was frozen in a dry ice/isopropanol bath and thawed in a 30°C bath. The sample was then extruded under 200-750 psi using two stacked 0.2 micron polycarbonate filter membranes (Nuclepore") in a 10 ml volume "Extruder" (Lipex Biomembranes Inc. , Vancouver, Canada) . The freeze/thaw and extrusion was repeated two more times. The final product was extruded two more times.
EXAMPLE 3: Encapsulation of FITC Dextran in Negatively Charged Liposomes
A quantity of 200 mg FITC Dextran (Sigma Chemical Co.) was dissolved in 1.5 ml 20 mM Tris buffer (pH 7.5) and the solution adjusted to 290 mOsm with NaCl. A lipid film, comprising 25 uMoles of total lipid, in a molar ratio of 4.9/2.1/3, DOPC/DOPG/Cholesterol, was prepared as in Example 2. The lipid film at 20βC was suspended in 1.5 ml of the FITC Dextran solution for 1 hour using a rotary shaker. The sample was frozen in a dry ice/isopropanol bath and thawed in a 30°C bath, then extruded under 200-750 PSI through stacked 0.2 micron polycarbonate filter membranes (Nuclepore'") in a 10 ml volume "Extruder" (Lipex Biomembranes Inc. , Vancouver, Canada) . The freeze/thaw and extrusion was repeated two more times. The final product was extruded two more times. The sample was applied to a Sephadex G-75 column equilibrated with the 20 mM tris buffer (adjusted to 290 mOsm with NaCl) and the void volume, which contains the liposome encapsulated FITC dextran, collected.
EXAMPLE 4 : Rhodamine-Phosphatidylethanolamine Labeled Negatively Charged Liposomes
A quantity of 22 mg egg phosphatidylcholine, 9.3 mg egg phosphatidylglycerol, 7.7 mg cholesterol and 1 mg of rhodamine-phosphatidylethanolamine were dissolved into 1 ml of chloroform. The lipid film was suspended at 20"C for 1 hour using a rotary shaker with Dulbecco's phosphate buffered saline (PBS) pH 7.4. The sample was frozen in a dry ice/isopropanol bath and thawed in a 30βC bath. The sample was extruded under 200-750 psi using two stacked 0.1 micron polycarbonate filter membranes (Nuclepore*) in a 10 ml volume "Extruder" (Lipex Biomembranes Inc., Vancouver,
Canada) . The freeze/thaw and extrusion was repeated two more times. The final product was extruded two more times.
EXAMPLE 5: Encapsulation of 6 carboxy-fluorescein in Negatively Charged Liposomes A quantity of 23.5 mg 6 carboxy-fluorescein (6CF) was placed into 2.5 ml of 20 mM phosphate buffer (pH 7.4); and approximately 160 ul of IN NaOH added to dissolve 6CF. The solution was readjusted to pH 7.4; and adjusted to 290 mOsm with NaCl. This solution contained 8.8 mg/ml 6CF (23.4 mM) . A quantity of 9.34 mg DOPC, 8.12 mg DOPG and 5.8 mg cholesterol was placed into 2 ml of chloroform, comprising 50 μM of total lipid in a ratio of 4.9/2.1/3 DOPC/DOPG/Cholesterol. The lipid film was suspended at 20βC for 1 hour using a rotary shaker with 0.75 ml of the 6CF solution. The sample was frozen in a dry ice/isopropanol bath and thawed in a 30"C bath. The sample was then extruded under 200-750 psi using two stacked 0.2 micron polycarbonate filter membranes (Nuclepore) in a 10 ml volume "Extruder" (Lipex Biomembranes Inc., Vancouver, Canada) . The freeze/thaw and extrusion was repeated two more times. The final product was extruded two more times. The sample was frozen in a dry ice isopropanol bath and thawed at room temperature. It was then extruded though 0.2 micron nucleopore filter membranes. The freeze/thaw and extrusion was repeated two more times. The final product was extruded two more times. The sample was applied to a Sephadex G-75 column equilibrated with the 20 mM tris buffer (adjusted to 290 mOsm with NaCl) and the void volume, containing the liposome encapsulated 6CF collected.
EXAMPLE 6: Encapsulation of Phalladion in Negatively Charged Liposomes
A volume of 3 ml of methanolic phalladion solution (3.3 μM) was dried on the Rotovap. The phalloidin peptide was dissolved into 0.5 ml of Tris buffer (20 mM pH 7.5) and adjusted to 290 mOsm) . A quantity of 4.67 mg DOPC, 4.06 mg DOPG and 2.9 mg cholesterol was taken up into 1 ml of chloroform. This results in 25 uMoles of total lipid, in a molar ratio of 4.9/2.1/1/3, DOPC/DOPG/Cholesterol. The lipid film was suspended at 20° C for 1 hour using a rotary shaker with 1.5 ml of the phalladion solution. The sample was frozen in a dry ice/isopropanol bath and thawed in a 30*C water bath. The sample was then extruded under 200-750 psi using two stacked 0.2 micron polycarbonate filter membranes (Nuclepore"') in a 10 ml volume "Extruder" (Lipex Biome branes Inc. , Vancouver, Canada) . The freeze/thaw and extrusion was repeated two more times. The dried lipid film was suspended in 3 ml of the phalloidin solution. The sample was frozen in a dry ice/isopropanol bath and thawed at room temperature. It was then extruded though 0.2 micron nucleopore filter membranes. The freeze/thaw and extrusion was repeated two more times. The final product was extruded two more times. The sample was then applied to a Sephadex G-75 column equilibrated with the 20 mM tris buffer (adjusted to 290 mOsm with NaCl) and the void volume, containing the liposome encapsulated phalladion collected.
EXAMPLE 7: Cells
PBS (Dulbecco s phosphate buffered saline without calcium or magnesium) , DMEM (Dulbecco's minimum essential medium) were obtained from Irvine Scientific (Irvine, CA.). Opti-MEM was obtained from Gibco Laboratories (Life Technologies, Inc. , Grand Island, New York) . Bovine calf serum was obtained from Hyclone. Trypsin, EDTA (0.5 gm trypsin, 0.2 gm EDTA/liter) in Hank's salts was from Irvine Scientific. NIH 3T3 cells, a contact inhibited mouse fibroblast line, are from Dr. Marguerite Vogt, Salk Institute. The cells are grown in monolayer in DMEM + 10% calf sera, 1% fungibact (Irvine Scientific) . Cells are removed by trypsinization with trypsin-EDTA solution. Cells are seeded onto 4 cm2 Lab-Tek tissue culture cells per well. NIH 3T3 cells are maintained in subconfluent stock cultures to retain their contact inhibition.
HELA cells are a human carcinoma epithelial cell line. The cells are maintained in DMEM + 10% fetal calf sera + 1% fungibact. The cells grow by attachment to a surface and trypsinized for transfer. Cells are seeded at low density for microscope slides at approximately 1000 cells per 4 cm2. EXAMPLE 8: Delivery of Detectable Substances into Cells Using Self-assembling Charged Lipid Complexes
Positively charged liposomes, prepared as described in
Example 1 were diluted with water to give a concentration of 1.15 ng/ul. Negatively charged liposomes, comprising
FITC dextran, 6-carboxy fluorescein, or phalladion, encapsulated in a negatively charged lipid vesicle and prepared according to Examples 4, 5 and 6 respectively, were diluted to give a concentration of approximately 0.05 ng/ul with Tris buffer (20 mM, pH 7.5; 290 mOsm) .
The substances were then delivered into either HeLa or 3T3 cells as follows:
Growing media was removed from the cells which were then washed with phosphate buffered saline (PBS) 2 times. Opti- Mem non-serum media (2 ml) was then to each 4 cm2 well. Quantities of 35 ul of empty positively charged lipid vesicles, prepared according to Example 1 and 80 ul of negatively charged lipid vesicles having the detectable substances indicated above encapsulated within were added to each of the wells containing cells. Plates were incubated for 2 hours at 37CC, then washed with PBS 2 times and viewed with the epi-fluorescent microscope using the appropriate filters.
RESULTS: Comparing phase contrast photomicrograph (a) with the corresponding fluorescence photomicrograph (b) in each case:
Figure 1 shows intracytoplasmic delivery of fluoresceinated dextran (MW 4000) encapsulated into negatively charged vesicles, into NIH-3T3 cells according to the methods previously described, using DOTMA/DOPE vesicles. This dye is considered to be a mimic for a water soluble peptide, and its behavior would be expected to be indicative of peptide delivery. Figure 2 gives similar results with HELA cells. A low molecular weight fluorescent dye, 6-carboxy fluorescein, encapsulated into negatively charged vesicles can also be delivered into 3T3 cells using DOTMA/DOPE by this method as indicated on Figure 4. Again the comparison with the light micrograph illustrated co-localization with the cytoplasm of the cells.
Figure 5 shows the results of delivery of negatively charged vesicle encapsulated Rhodamine-phalladion, into NIH-3T3 cells using DOTAP/DOPE. Fluorescence is confined to the cytoplasm in these micrographs and the pattern of fluorescence is typical of phalladion binding to intracellular actin filaments.
Similarly the lipophilic dye, Rhodamine-PE, prepared as indicated in Example 6, can also be delivered intracellularly by this method, into NIH-3T3 cells using DOTMA/PE vesicles (Figure 3) . Comparison with the light photomicrograph indicates a concentration of fluorescence intracellularly.
EXAMPLE 9: A Kit for Intracellular Delivery Procedures
A kit was constructed to provide materials to accomplish the delivery and uptake of macromolecules into tissue culture cells. The materials in the kit consisted of a vial containing dried lipid, a buffer solution, a solution containing cationic lipid vesicles, a syringe provided with a 0.4 micron Nuclepore™ membrane in a filter housing, and an instruction leaflet. The lipid vial contained 35 mg of l-palmitoyl-2-oleoyl-phosphatidylcholine and 15 mg of dioleoylphosphatidylglycerol in a dried lipid film. The buffer solution consisted of 130 mM NaCl and 10 mM sodium phosphate buffer at a pH of 7.4.
The kit was used to deliver monoclonal antibodies to intercellular actin for transfection into mammalian cells according to the instructions provided. A quantity of the macromolecule to be delivered, in this case, 1 mg of the actin antibody, was delivered into the buffer solution and mixed well. The solution was then placed in the lipid- containing vial to hydrate the lipid film and form a liposome suspension. The lipid suspension was then loaded into the barrel of the syringe provided, and the liposome suspension was forced out through the filter membrane to form smaller lipid vesicles of a more uniform size. Under these conditions approximately 10% of the initial quantity of macromolecule is encapsulated into the lipid vesicles.
The cationic lipid vesicle solution provided in the kit consisted of 0.5 mg/ml of a DORI diether and 0.5 mg/ml dioleoylphosphatidylethanolamine. The user is instructed to combine 5μl of the negatively charged liposomes with 100 μl of the DORI/DOPE solution and to add the mixture onto tissue culture cells (approximately 106 cells) growing on OptiMem™ media without serum. The incubation of the cells then continues for 2 hours. The cells are washed, and then assayed for the biological endpoint, as in Example 8.
There will be various modifications, improvements, and applications of the disclosed invention that will be apparent to those of skill in the art, and the present application is intended to cover such embodiments. Although the present invention has been described in the context of certain preferred embodiments, it is intended that the full scope of the disclosure be measured by reference to the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method for making a vehicle for administering a bioactive substance to a cell, comprising the steps of: providing said substance in a first lipid vesicle, said vesicle comprising at least one negatively charged lipid and having a net negative charge; and combining said first lipid vesicle with second lipid vesicles, said vesicles comprising at least one cationic lipid and having a net positive charge; whereby said first lipid vesicles are coated with at least one positively charged lipid vesicle.
2. The method of Claim 1, wherein said bioactive substance is lipophilic or comprises lipophilic regions, and is thereby incorporated into the lipid structure of said first lipid vesicle.
3. The method of Claim 1, wherein said bioactive substance is encapsulated within said first lipid vesicle.
4. A method for making a vehicle for administering a bioactive substance to a cell, comprising the steps of: contacting a bioactive substance with at least one lipid vesicle, said lipid vesicle comprising a negatively charged lipid species and having a net negative charge, whereby said bioactive substance and said lipid vesicle form a first lipid complex; and contacting said first lipid complex with lipid vesicles comprising at least one cationic lipid and having a net positive charge, whereby said first lipid complex and said positively charged lipid vesicles form a second lipid complex.
5. The method of Claim 4 wherein said bioactive substance is lipophilic or has lipophilic regions.
6. The method of Claim 4, wherein said bioactive substance has positively charged regions or has a net positive charge.
7. The method of any one of Claims 1 through 6, further comprising the steps of:
(a) contacting said vehicle with negatively charged lipid vesicles to form a lipid complex having negatively charged regions on its outer surface; and
(b) contacting said lipid complex with positively charged lipid vesicles to form a vehicle having positively charged regions on its outer surface.
8. The method of Claim 7, further comprising repeating steps (a) and (b) at least once.
9. A method for administering a bioactive substance to a cell, comprising the steps of: providing said substance in a negatively charged lipid vesicle; administering said negatively charged complex to a cell together with lipid vesicles comprising at least one cationic lipid, said vesicles having a net positive charge.
10. A method for administering a bioactive substance to a cell, comprising the steps of: preparing a complex comprising said bioactive substance and a negatively charged lipid, said complex having a net negative charge; administering said negatively charged complex to a cell together with lipid vesicles comprising at least one cationic lipid, said vesicles having a net positive charge.
11. The method of Claim 9 or 10 wherein said bioactive substance is administered in vivo.
12. The method of any one of Claims 1 through 6 wherein said cationic lipid has the structure
Figure imgf000038_0001
wherein
Y1 and Y2 are the same or different and are -0-CH2-, -O-C(O)-, or -0- ;
R1 and R2 are the same or different and are H, C^ to C23 alkyl or alkenyl;
R3 and R4 are the same or different and are C^ to C24 alkyl, or H;
R5 is O to C24 alkyl straight chain or branched chain;
R6 is -C(0)-(CH2)m-NH-, a diaminocarboxylic acid which is alkyl, aryl, or aralkyl, or -C(0)-(CH2)m"NH- linked to said diaminocarboxylic acid, or is absent;
R7 is H, spermine, spermidine, a histone, or a protein with DNA-binding specificity, or the same groups wherein the amines of the R7 moiety are quaternized with R3, R4, or R5 groups; or
R7 is a L- or D-alpha amino acid having a positively charged group on the side chain, said amino acids comprising arginine, histidine, lysine or ornithine or derivatives thereof, or the same amino acids wherein the amine of the R7 moiety is quaternized with R3, R4 or R5 groups; or
R7 is a polypeptide selected from the group consisting of L- or D-alpha amino acids, wherein at least one of the amino acids residues comprises arginine, histidine, lysine, ornithine, or derivatives thereof; n is l to 8; m is 1 to 18; and X is a non-toxic anion, providing that when n is 1,
R5 is -CH2-CH2-, R6 is absent, and R7 is H, then R1 and R2 are not both stearoyl.
13. A method according to Claim 12 wherein said cationic lipid is DL-l,2-dioleoyl-3-dimethylaminopropyl- - hydroxyethylammonium and its salts.
14. A method according to Claim 12 wherein Y1 and Y2 are different and are either -0-CH2- or -O-C(O)-.
15. A method according to Claim 14 wherein at least on e c at i o n i c l i p id i s 1-O-ol eyl- , 2-oleoyl-3-dimethylaminopropyl- -hydroxyethylammonium.
16. A method according to any one of Claims 1 through 6 , where in s a i d cat i oni c l ipid is l,2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP) .
17. A method according to any one of Claims 1 through 6 wherein said cationic lipid is an
N-(ω, ω-1-dialkoxy) -alk-l-yl-N, N, N-trisubstituted ammonium surfactant.
18. A method according to Claims 1 through 6 wherein at least one cationic lipid has the structure
Figure imgf000039_0001
or an optical isomer thereof, wherein
R1 and R2 are individually absent, Ci to C23 alkyl or alkenyl, or H;
Y1 and Y2 are different and are either -0-CH2-, -0-C(0)- or
OH;
R3, R4 and R5 are the same or different and are H, C^ to
C14 alkyl, C7 to Cn aryl or alkaryl, or at least two of R3, R4 and R5 are taken together to form quinuclidino, piperidino, pyrrolidino, or orpholino; n is 1 to 22; and
X is a non-toxic anion.
19. A composition of matter, comprising: a bioactive substance contained in at least one negatively charged lipid vesicle; and lipid vesicles comprising at least one cationic lipid species and having a net positive charge at least partially covering said negatively charged lipid vesicle.
20. The composition of Claim 19 wherein said bioactive substance is encapsulated within said negatively charged lipid vesicle.
21. The composition of Claim 19 comprising a bioactive substance which is lipophilic or has a lipophilic region whereby said bioactive substance is incorporated among the lipids of said negatively charged lipid vesicle.
22. A composition of matter comprising a primary complex of a bioactive substance and at least one negatively charged lipid vesicle; and lipid vesicles comprising at least one cationic lipid species and having a net positive charge at least partially covering said negatively charged lipid vesicle to form a secondary complex.
23. The composition of Claim 22 wherein said bioactive substance comprises positively charged regions or has a net positive charge.
24. The composition of Claim 19 or 22, wherein εaid composition has a net positive charge.
25. The composition of Claim 19 or 22 wherein said bioactive substance is a protein or polypeptide.
26. The composition of Claim 19 or 22 wherein said bioactive substance is a polynucleotide.
27. The composition of Claim 26 wherein said bioactive substance is an antisense polynucleotide.
28. The composition of Claim 19 or 22 wherein said bioactive substance is a drug.
29. The composition of any one of Claims 19 through
27 wherein at least one cationic lipid has the structure
Figure imgf000041_0001
wherein Y1 and Y2 are the same or different and are -0-CH2-,-
O-C(O)-, or OH;
R1 and R2 are the same or different and are H, C^ to C23 alkyl or alkenyl, or are absent;
R3 and R4 are the same or different and are Ci to C24 alkyl, or H;
R5 is Ci to C24 alkyl straight chain or branched chain;
R6 is -C(0)-(CH2)m-NH-( a diaminocarboxylic acid which is alkyl, aryl, or aralkyl, or -C(O)-(CH2)m-NH- linked to said diaminocarboxylic acid, or is absent; R7 is H, spermine, sperinidine or a histone or the same groups wherein the amine is quaternized with R3, R4, or R5 groups; or
R7 is an L- or D-alpha amino acid having a positively charged group on the side chain, said amino acids comprising arginine, histidine, lysine or ornithine or the same amino acids wherein the amine is quaternized with R3,
R4 or R5 groups, or polypeptides comprising said L- or D- alpha amino acids; n is 1 to 8; m is 1 to 18; and
X is a non-toxic anion, providing that when n is l,
R5 is -CH2-CH2-, R6 is absent, and R7 is H, then R1 and R2 are not both stearoyl.
30. The composition of Claim 29, wherein at least one cationic lipid is 1,2-dioleoy1-3-dimethylaminopropy1-/3-hydroxyethylammonium.
31. The composition of Claim 29, wherein Y1 and Y2 are different and are either -0-CH2- or -0-C(0)-.
32. The composition of Claim 31, wherein at least one cationic lipid is 1-O-oleyl-,2-oleoyl-3-dimethylaminopropyl
-β-hydroxyethylammoniurn.
33. The composition of Claim 29, wherein said cationic lipid is 1,2-bis(oleoyloxy) 3-(trimethylammonio)- propane (DOTAP) .
34. The composition of Claim 29, wherein said cationic lipid is an N-(ω, ω-1-dialkoxy)-alk-1-yl-N, N, N-trisubstituted ammonium surfactant.
35. The composition of Claim 29, wherein at least one cationic lipid has the structure
(II)
Figure imgf000042_0002
Figure imgf000042_0001
or an optical isomer thereof, wherein
R1 and R2 are individually absent, C to C23 alkyl or alkenyl;
Y1 and Y2 are different and are either -0-CH2-, -O-C(O)- or OH;
R3, R4 and R5 are the same or different and are H, C^ to
C14 alkyl, C7 to Cn aryl or alkaryl, or at least two of
R3, R4 and R5 are taken together to form quinuclidino, piperidino, pyrrolidino, or morpholino; n is 1 to 22; and
X is a non-toxic anion.
36. A kit for preparing self-assembling lipid complexes for the delivery of a bioactive substance to a cell, either in vi tro or i?> vivo , comprising: at least one vessel containing at least one negatively charged lipid species; a quantity of a lipid substance, said substance comprising at least one cationic lipid species.
37. The kit of Claim 36, further comprising at leaεt one hydrating buffer solution.
38. The kit of Claim 36, further comprising a membrane filter device.
39. The kit of Claim 36, wherein said cationic lipid species has the structure
Figure imgf000043_0001
wherein
Y1 and Y2 are the same or different and are -0-CH2-/- O-C(O)-, or OH;
R1 and R2 are the same or different and are H, C^ to C23 alkyl or alkenyl, or are absent;
R3 and R4 are the same or different and are 0 to C24 alkyl, or H;
R5 is Ci to C24 alkyl straight chain or branched chain; R6 is -C(0)-(CH2)m-NH-, a diaminocarboxylic acid which is alkyl, aryl, or aralkyl, or -C(0)-(CH2)m-NH- linked to said diaminocarboxylic acid, or is absent;
R7 is H, εpermine, spermidine or a histone or the same groups wherein the amine is quaternized with R3, R4, or R5 groups; or R7 is an L- or D-alpha amino acids having a positively charged group on the side chain, said amino acids comprising arginine, histidine, lysine or ornithine or the same amino acids wherein the amine is quaternized with R3, R4 or R5 groups, or polypeptides comprising said L- or D- alpha amino acids; n is l to 8; m is 1 to 18; and
X is a non-toxic anion, providing that when n is l,
R5 is -CH2-CH2-, R6 is absent, and R7 is H, then R1 and R2 are not both stearoyl.
PCT/US1991/002962 1990-05-03 1991-04-29 Intracellular delivery of biologically active substances by means of self-assembling lipid complexes WO1991017424A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US51929190A 1990-05-03 1990-05-03
US519,291 1990-05-03

Publications (1)

Publication Number Publication Date
WO1991017424A1 true WO1991017424A1 (en) 1991-11-14

Family

ID=24067677

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1991/002962 WO1991017424A1 (en) 1990-05-03 1991-04-29 Intracellular delivery of biologically active substances by means of self-assembling lipid complexes

Country Status (2)

Country Link
AU (1) AU7979491A (en)
WO (1) WO1991017424A1 (en)

Cited By (394)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0523189A1 (en) * 1990-04-19 1993-01-20 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
WO1997019675A2 (en) * 1995-11-30 1997-06-05 Vical Incorporated Complex cationic lipids
US5667764A (en) * 1988-05-02 1997-09-16 Zynaxis, Inc. Compounds, compositions and methods for binding bio-affecting substances to surface membranes of bio-particles
US5693769A (en) * 1991-12-13 1997-12-02 Transcell Technologies, Inc. Glycosylated steroid derivatives for transport across biological membranes and process for making and using same
WO1998010748A1 (en) * 1996-09-13 1998-03-19 The School Of Pharmacy Liposomes
WO1998014593A2 (en) 1996-10-01 1998-04-09 Geron Corporation Human telomerase catalytic subunit
US5770580A (en) * 1992-04-13 1998-06-23 Baylor College Of Medicine Somatic gene therapy to cells associated with fluid spaces
US5795870A (en) * 1991-12-13 1998-08-18 Trustees Of Princeton University Compositions and methods for cell transformation
FR2766706A1 (en) * 1997-07-30 1999-02-05 Biovector Therapeutics Sa STABLE, NEUTRAL OR NEGATIVE GLOBAL PARTICLE COMPLEXES OF A MULTILAMELLAR STRUCTURE COMPRISING AT LEAST ONE GLOBALLY ANIONIC BIOLOGICALLY ACTIVE SUBSTANCE AND A CATIONIC COMPONENT, THEIR PREPARATION AND USE
FR2766705A1 (en) * 1997-07-30 1999-02-05 Biovector Therapeutics Stable particulate complexes for use in gene therapy
US5928944A (en) * 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection
US6057299A (en) * 1994-01-13 2000-05-02 Calydon, Inc. Tissue-specific enhancer active in prostate
US6136792A (en) * 1994-01-13 2000-10-24 Calydon, Inc. Prostate specific enhancer polynucleotides and methods of use thereof
WO2000063366A2 (en) * 1999-04-20 2000-10-26 Sequitur, Inc. Improved antisense oligomers
US6139871A (en) * 1995-07-26 2000-10-31 The University Of British Columbia Liposome compositions and methods for the treatment of atherosclerosis
US6174871B1 (en) 1995-02-28 2001-01-16 The Regents Of The University Of California Gene therapies for enhancing cardiac function
US6245750B1 (en) 1998-01-23 2001-06-12 Newbiotics, Inc. Enzyme catalyzed therapeutic agents
US6312719B1 (en) 1994-03-04 2001-11-06 The University Of British Columbia Liposome compositions and methods for the treatment of atherosclerosis
US6367479B1 (en) 1995-10-11 2002-04-09 Esperion Luv Development, Inc. Method of forcing the reverse transport of cholesterol from a body part to the liver while avoiding harmful disruptions of hepatic cholesterol homeostasis, and pharmaceutical compositions and kit related thereto
US6683061B1 (en) 1999-07-22 2004-01-27 Newbiotics, Inc. Enzyme catalyzed therapeutic activation
US6716882B2 (en) 1993-12-20 2004-04-06 Invitrogen Corporation Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US6752987B1 (en) 1995-02-28 2004-06-22 The Regents Of The University Of California Adenovirus encoding human adenylylcyclase (AC) VI
US6773719B2 (en) 1994-03-04 2004-08-10 Esperion Luv Development, Inc. Liposomal compositions, and methods of using liposomal compositions to treat dislipidemias
DE10322123A1 (en) * 2003-05-12 2004-12-16 Novosom Ag Stable anionic liposome depot system for prolonged release of protein or peptide drugs, e.g. insulin, comprising phosphatidyl choline, cholesterol, anionic lipid(s) and cationic polymer
WO2005014791A2 (en) 2003-08-08 2005-02-17 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
US6890554B2 (en) 1993-06-01 2005-05-10 Invitrogen Corporation Genetic immunization with cationic lipids
US6916918B2 (en) 1997-08-04 2005-07-12 Cell Genesys, Inc. Human glandular kallikrein enhancer, vectors comprising the enhancer and methods of use thereof
US6989434B1 (en) 1994-02-11 2006-01-24 Invitrogen Corporation Reagents for intracellular delivery of macromolecules
US6995008B1 (en) 1994-03-07 2006-02-07 Merck & Co., Inc. Coordinate in vivo gene expression
US7094756B2 (en) 2001-06-22 2006-08-22 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Abin-mediated hepatitis protection
US7122656B2 (en) 2002-01-10 2006-10-17 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Splice variant of MyD88 and uses thereof
US7135324B2 (en) 2002-09-04 2006-11-14 The University Of Connecticut Viral recombinases, related articles, and methods of use thereof
US7138388B2 (en) 2001-01-19 2006-11-21 Celmed Oncology (Usa), Inc. Methods to treat autoimmune and inflammatory conditions
WO2006130974A1 (en) 2005-06-08 2006-12-14 Cangene Corporation Hyaluronic acid binding peptides enhance host defense against pathogenic bacteria
WO2007062399A2 (en) 2005-11-23 2007-05-31 The Board Of Regents Of The University Of Texas System Oncogenic ras-specific cytotoxic compound and methods of use thereof
US7235236B2 (en) 1995-02-28 2007-06-26 The Regents Of The University Of California Polynucleotide encoding human adenylylcyclase VI and uses thereof for enhancing cardiac function
US7244450B2 (en) 1999-04-01 2007-07-17 Inex Pharmaceuticals Corporation Compositions and methods for treating lymphoma
US7247316B2 (en) 1999-04-01 2007-07-24 Board Of Regents, The University Of Texas System Compositions and methods for treating lymphoma
US7273620B1 (en) 2002-05-20 2007-09-25 University Of British Columbia Triggered release of liposomal drugs following mixing of cationic and anionic liposomes
US7311924B2 (en) 1999-04-01 2007-12-25 Hana Biosciences, Inc. Compositions and methods for treating cancer
US7358085B2 (en) 2005-02-28 2008-04-15 Sangamo Biosciences, Inc. Anti-angiogenic methods and compositions
US7384923B2 (en) 1999-05-14 2008-06-10 Lipoxen Technologies Limited Liposomes
US7407776B2 (en) 2003-09-19 2008-08-05 Sangamo Biosciences, Inc. Engineered zinc finger proteins for regulation of gene expression
US7414026B2 (en) 1998-05-06 2008-08-19 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Inhibitors of NF-kappaB activation
US7419968B1 (en) 1999-07-22 2008-09-02 Celmed Oncology (Usa), Inc. Methods for treating therapy-resistant tumors
EP1985285A2 (en) 1999-04-01 2008-10-29 Hana Biosciences, Inc. Compositions and methods for treating lymphoma
US7462605B2 (en) 1998-01-23 2008-12-09 Celmed Oncology (Usa), Inc. Phosphoramidate compounds and methods of use
US7534775B2 (en) 2004-04-08 2009-05-19 Sangamo Biosciences, Inc. Methods and compositions for modulating cardiac contractility
EP2128248A1 (en) 2002-02-01 2009-12-02 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
WO2009151591A2 (en) 2008-06-10 2009-12-17 Sangamo Biosciences, Inc. Methods and compositions for generation of bax- and bak-deficient cell lines
WO2010021692A1 (en) 2008-08-22 2010-02-25 Sangamo Biosciences, Inc. Methods and compositions for targeted single-stranded cleavage and targeted integration
WO2010065123A1 (en) 2008-12-04 2010-06-10 Sangamo Biosciences, Inc. Genome editing in rats using zinc-finger nucleases
EP2207032A1 (en) 2000-02-08 2010-07-14 Sangamo BioSciences, Inc. Cells expressing zinc finger protein for drug discovery
EP2213731A1 (en) 2006-05-25 2010-08-04 Sangamo BioSciences, Inc. Variant foki cleavage half-domains
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US7811602B2 (en) 2004-05-17 2010-10-12 Tekmira Pharmaceuticals Corporation Liposomal formulations comprising dihydrosphingomyelin and methods of use thereof
WO2010117464A1 (en) 2009-04-09 2010-10-14 Sangamo Biosciences, Inc. Targeted integration into stem cells
WO2010148010A1 (en) 2009-06-15 2010-12-23 4S3 Bioscience Inc. Methods and compositions for treatment of myotubular myopathy using chimeric polypeptides comprising myotubularih 1 (mtm1) polypeptides
EP2266396A2 (en) 2001-09-24 2010-12-29 Sangamo BioSciences, Inc. Modulation of stem cells using zinc finger proteins
US7868162B2 (en) 1998-12-30 2011-01-11 Lakewood-Amedex, Inc. Antimicrobial and antiviral compounds and methods for their use
EP2292274A1 (en) 2004-09-16 2011-03-09 Sangamo BioSciences, Inc. Compositions and methods for protein production
WO2011043980A1 (en) 2009-10-07 2011-04-14 Sanford Burnham Medical Research Institute Methods and compositions related to clot-binding lipid compounds
EP2311466A2 (en) 2001-05-03 2011-04-20 Lakewood-Amedex, Inc Antimicrobial compounds and methods for their use
EP2345417A1 (en) 2005-12-22 2011-07-20 Vib Vzw Means and methods for mediating protein interference
WO2011090804A1 (en) 2010-01-22 2011-07-28 Dow Agrosciences Llc Targeted genomic alteration
EP2351765A2 (en) 2001-07-10 2011-08-03 Lakewood-Amedex, Inc Oligonucleotide-containing pharmacological compositions and their use
WO2011100058A1 (en) 2010-02-09 2011-08-18 Sangamo Biosciences, Inc. Targeted genomic modification with partially single-stranded donor molecules
WO2011119871A1 (en) 2010-03-24 2011-09-29 Rxi Phrmaceuticals Corporation Rna interference in ocular indications
WO2011119887A1 (en) 2010-03-24 2011-09-29 Rxi Pharmaceuticals Corporation Rna interference in dermal and fibrotic indications
WO2011130729A2 (en) 2010-04-16 2011-10-20 Salk Institute For Biological Studies Methods for treating metabolic disorders using fgf
EP2397549A2 (en) 1999-02-04 2011-12-21 BP Corporation North America Inc. Non-stochastic generation of genetic vaccines and enzymes
WO2012047598A1 (en) 2010-09-27 2012-04-12 Sangamo Biosciences, Inc. Methods and compositions for inhibiting viral entry into cells
US8158595B2 (en) 2006-11-09 2012-04-17 California Institute Of Technology Modular aptamer-regulated ribozymes
WO2012061113A2 (en) 2010-10-25 2012-05-10 Academia Sinica, Taiwan Cancer-targeting peptides and uses thereof in cancer treatment and diagnosis
US8313925B2 (en) 2005-07-26 2012-11-20 Sangamo Biosciences, Inc. Zinc finger proteins and method for inactivating a dhfr gene in a chinese hamster ovary cell
EP2527439A2 (en) 2007-12-11 2012-11-28 The Scripps Research Institute Compositions and methods related to mRNA translational enhancer elements
EP2527435A2 (en) 2007-07-12 2012-11-28 Sangamo BioSciences, Inc. Methods and compositions for inactivating alpha 1,6 fucosyltransferase (FUT 8) gene expression
US8329882B2 (en) 2009-02-18 2012-12-11 California Institute Of Technology Genetic control of mammalian cells with synthetic RNA regulatory systems
WO2013012674A1 (en) 2011-07-15 2013-01-24 The General Hospital Corporation Methods of transcription activator like effector assembly
US8367815B2 (en) 2007-08-28 2013-02-05 California Institute Of Technology Modular polynucleotides for ligand-controlled regulatory systems
US8435960B2 (en) 1998-12-30 2013-05-07 Lakewood-Amedex, Inc. Devices for improved wound management
EP2615106A1 (en) 2010-02-08 2013-07-17 Sangamo BioSciences, Inc. Engineered cleavage half-domains
WO2013130824A1 (en) 2012-02-29 2013-09-06 Sangamo Biosciences, Inc. Methods and compositions for treating huntington's disease
WO2014011237A1 (en) 2012-07-11 2014-01-16 Sangamo Biosciences, Inc. Methods and compositions for the treatment of lysosomal storage diseases
WO2014011901A2 (en) 2012-07-11 2014-01-16 Sangamo Biosciences, Inc. Methods and compositions for delivery of biologics
US8664189B2 (en) 2008-09-22 2014-03-04 Rxi Pharmaceuticals Corporation RNA interference in skin indications
WO2014036219A2 (en) 2012-08-29 2014-03-06 Sangamo Biosciences, Inc. Methods and compositions for treatment of a genetic condition
US8669418B2 (en) 2005-12-22 2014-03-11 Vib Vzw Means and methods for mediating protein interference
WO2014059173A2 (en) 2012-10-10 2014-04-17 Sangamo Biosciences, Inc. T cell modifying compounds and uses thereof
WO2014059255A1 (en) 2012-10-12 2014-04-17 The General Hospital Corporation Transcription activator-like effector (tale) - lysine-specific demethylase 1 (lsd1) fusion proteins
EP2727600A1 (en) 2009-07-28 2014-05-07 Sangamo BioSciences, Inc. Methods and compositions for treating trinucleotide repeat disorders
WO2014089541A2 (en) 2012-12-07 2014-06-12 Haplomics, Inc. Factor viii mutation repair and tolerance induction
WO2014093622A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
WO2014093635A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
WO2014093661A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
WO2014093694A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
WO2014093595A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
WO2014093655A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
WO2014093712A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2014093701A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
WO2014130723A1 (en) 2013-02-20 2014-08-28 Valerion Therapeutics, Llc Methods and compositions for treatment of pompe disease
EP2784162A1 (en) 2012-12-12 2014-10-01 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US8865667B2 (en) 2007-09-12 2014-10-21 California Institute Of Technology Higher-order cellular information processing devices
WO2014190181A1 (en) 2013-05-22 2014-11-27 Northwestern University Rna-directed dna cleavage and gene editing by cas9 enzyme from neisseria meningitidis
WO2014197840A1 (en) 2013-06-07 2014-12-11 Massachusetts Institute Of Technology Affinity-based detection of ligand-encoded synthetic biomarkers
WO2014204728A1 (en) 2013-06-17 2014-12-24 The Broad Institute Inc. Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of post mitotic cells
WO2014204729A1 (en) 2013-06-17 2014-12-24 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using viral components
US8956646B2 (en) 2010-08-14 2015-02-17 The Regents Of The University Of California Zwitterionic lipids
WO2015031619A1 (en) 2013-08-28 2015-03-05 Sangamo Biosciences, Inc. Compositions for linking dna-binding domains and cleavage domains
WO2015057980A1 (en) 2013-10-17 2015-04-23 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
US9029524B2 (en) 2007-12-10 2015-05-12 California Institute Of Technology Signal activated RNA interference
WO2015070212A1 (en) 2013-11-11 2015-05-14 Sangamo Biosciences, Inc. Methods and compositions for treating huntington's disease
US9040495B2 (en) 2007-08-28 2015-05-26 California Institute Of Technology General composition framework for ligand-controlled RNA regulatory systems
WO2015085113A1 (en) 2013-12-04 2015-06-11 Rxi Pharmaceuticals Corporation Methods for treatment of wound healing utilizing chemically modified oligonucleotides
US9057057B2 (en) 2010-07-27 2015-06-16 The Johns Hopkins University Obligate heterodimer variants of foki cleavage domain
WO2015089486A2 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Systems, methods and compositions for sequence manipulation with optimized functional crispr-cas systems
WO2015089419A2 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components
WO2015089465A1 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for hbv and viral diseases and disorders
US9074224B2 (en) 2009-08-03 2015-07-07 Recombinetics, Inc. Methods and compositions for targeted gene modification
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
WO2015117081A2 (en) 2014-02-03 2015-08-06 Sangamo Biosciences, Inc. Methods and compositions for treatment of a beta thalessemia
WO2015143046A2 (en) 2014-03-18 2015-09-24 Sangamo Biosciences, Inc. Methods and compositions for regulation of zinc finger protein expression
US9145555B2 (en) 2009-04-02 2015-09-29 California Institute Of Technology Integrated—ligand-responsive microRNAs
WO2015168605A1 (en) 2014-05-01 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treatment of disorders in the front of the eye utilizing nucleic acid molecules
WO2015168108A2 (en) 2014-04-28 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treating cancer using nucleic targeting mdm2 or mycn
WO2015171932A1 (en) 2014-05-08 2015-11-12 Sangamo Biosciences, Inc. Methods and compositions for treating huntington's disease
EP2947146A1 (en) 2004-02-05 2015-11-25 Sangamo BioSciences, Inc. Methods and compositions for targeted cleavage and recombination
WO2015184268A1 (en) 2014-05-30 2015-12-03 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods of delivering treatments for latent viral infections
EP2980220A1 (en) 2005-09-20 2016-02-03 BASF Plant Science GmbH Improved methods controlling gene expression
WO2016037071A2 (en) 2014-09-05 2016-03-10 Rxi Pharmaceuticals Corporation Methods for treating aging and skin disorders using nucleic acids targeting tyr or mmp1
WO2016044416A1 (en) 2014-09-16 2016-03-24 Sangamo Biosciences, Inc. Methods and compositions for nuclease-mediated genome engineering and correction in hematopoietic stem cells
WO2016049163A2 (en) 2014-09-24 2016-03-31 The Broad Institute Inc. Use and production of chd8+/- transgenic animals with behavioral phenotypes characteristic of autism spectrum disorder
WO2016049251A1 (en) 2014-09-24 2016-03-31 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for modeling mutations in leukocytes
US9309568B2 (en) 2004-10-05 2016-04-12 California Institute Of Technology Aptamer regulated nucleic acids and uses thereof
EP3009511A2 (en) 2015-06-18 2016-04-20 The Broad Institute, Inc. Novel crispr enzymes and systems
US9358300B2 (en) 1998-11-12 2016-06-07 Life Technologies Corporation Transfection reagents
WO2016094874A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Escorted and functionalized guides for crispr-cas systems
WO2016094872A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Dead guides for crispr transcription factors
WO2016094867A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Protected guide rnas (pgrnas)
WO2016106244A1 (en) 2014-12-24 2016-06-30 The Broad Institute Inc. Crispr having or associated with destabilization domains
WO2016106236A1 (en) 2014-12-23 2016-06-30 The Broad Institute Inc. Rna-targeting system
WO2016108926A1 (en) 2014-12-30 2016-07-07 The Broad Institute Inc. Crispr mediated in vivo modeling and genetic screening of tumor growth and metastasis
WO2016123230A1 (en) 2015-01-28 2016-08-04 Pioneer Hi-Bred International, Inc. Crispr hybrid dna/rna polynucleotides and methods of use
EP3061834A1 (en) 2005-10-21 2016-08-31 The Regents of the University of California Shp-2 gene mutations in melanoma
WO2016161446A1 (en) 2015-04-03 2016-10-06 Dana-Farber Cancer Institute, Inc. Composition and methods of genome editing of b-cells
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
WO2016186772A2 (en) 2015-05-16 2016-11-24 Genzyme Corporation Gene editing of deep intronic mutations
WO2016196388A1 (en) 2015-05-29 2016-12-08 Juno Therapeutics, Inc. Composition and methods for regulating inhibitory interactions in genetically engineered cells
WO2016205764A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Novel crispr enzymes and systems
WO2016205728A1 (en) 2015-06-17 2016-12-22 Massachusetts Institute Of Technology Crispr mediated recording of cellular events
WO2016205749A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Novel crispr enzymes and systems
WO2017009489A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Topical compositions
WO2017009488A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Topical compositions
WO2017009491A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Peptides for use in promoting transport of glucose
WO2017011519A1 (en) 2015-07-13 2017-01-19 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
WO2017009492A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Antibacterial peptides, and uses thereof
WO2017024317A2 (en) 2015-08-06 2017-02-09 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
US9592250B2 (en) 2002-02-01 2017-03-14 Life Technologies Corporation Double-stranded oligonucleotides
EP3156062A1 (en) 2010-05-17 2017-04-19 Sangamo BioSciences, Inc. Novel dna-binding proteins and uses thereof
WO2017070605A1 (en) 2015-10-22 2017-04-27 The Broad Institute Inc. Type vi-b crispr enzymes and systems
WO2017070151A1 (en) 2015-10-19 2017-04-27 Rxi Pharmaceuticals Corporation Reduced size self-delivering nucleic acid compounds targeting long non-coding rna
WO2017106657A1 (en) 2015-12-18 2017-06-22 The Broad Institute Inc. Novel crispr enzymes and systems
WO2017106537A2 (en) 2015-12-18 2017-06-22 Sangamo Biosciences, Inc. Targeted disruption of the mhc cell receptor
WO2017106528A2 (en) 2015-12-18 2017-06-22 Sangamo Biosciences, Inc. Targeted disruption of the t cell receptor
WO2017123757A1 (en) 2016-01-15 2017-07-20 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of neurologic disease
WO2017165788A1 (en) 2016-03-24 2017-09-28 Marquette University Quantitative flagellar fluorescent markers and standards
US20170298297A1 (en) * 2014-10-01 2017-10-19 Yeda Research And Development Co. Ltd. Liposomes-containing antifouling compositions and uses thereof
WO2017184786A1 (en) 2016-04-19 2017-10-26 The Broad Institute Inc. Cpf1 complexes with reduced indel activity
WO2017184768A1 (en) 2016-04-19 2017-10-26 The Broad Institute Inc. Novel crispr enzymes and systems
US9801874B2 (en) 2012-11-20 2017-10-31 Spectrum Pharmaceuticals Method for the preparation of liposome encapsulated vincristine for therapeutic use
WO2017189308A1 (en) 2016-04-19 2017-11-02 The Broad Institute Inc. Novel crispr enzymes and systems
WO2017219027A1 (en) 2016-06-17 2017-12-21 The Broad Institute Inc. Type vi crispr orthologs and systems
WO2018005873A1 (en) 2016-06-29 2018-01-04 The Broad Institute Inc. Crispr-cas systems having destabilization domain
WO2018013932A1 (en) 2016-07-15 2018-01-18 Salk Institute For Biological Studies Methods and compositions for genome editing in non-dividing cells
WO2018013840A1 (en) 2016-07-13 2018-01-18 Vertex Pharmaceuticals Incorporated Methods, compositions and kits for increasing genome editing efficiency
WO2018014936A1 (en) 2016-07-18 2018-01-25 Nuritas Limited Topical compositions
US9890364B2 (en) 2012-05-29 2018-02-13 The General Hospital Corporation TAL-Tet1 fusion proteins and methods of use thereof
WO2018029586A1 (en) 2016-08-07 2018-02-15 Novartis Ag Mrna-mediated immunization methods
WO2018035388A1 (en) 2016-08-17 2018-02-22 The Broad Institute, Inc. Novel crispr enzymes and systems
WO2018035387A1 (en) 2016-08-17 2018-02-22 The Broad Institute, Inc. Novel crispr enzymes and systems
WO2018039440A1 (en) 2016-08-24 2018-03-01 Sangamo Therapeutics, Inc. Regulation of gene expression using engineered nucleases
WO2018039448A1 (en) 2016-08-24 2018-03-01 Sangamo Therapeutics, Inc. Engineered target specific nucleases
WO2018067826A1 (en) 2016-10-05 2018-04-12 Cellular Dynamics International, Inc. Generating mature lineages from induced pluripotent stem cells with mecp2 disruption
WO2018071873A2 (en) 2016-10-13 2018-04-19 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
EP3311822A1 (en) 2010-11-17 2018-04-25 Sangamo Therapeutics, Inc. Methods and compositions for modulating pd1
WO2018081775A1 (en) 2016-10-31 2018-05-03 Sangamo Therapeutics, Inc. Gene correction of scid-related genes in hematopoietic stem and progenitor cells
EP3329930A1 (en) 2016-12-05 2018-06-06 Nuritas Limited Pharmaceuctical compositions
EP3329905A1 (en) 2016-12-05 2018-06-06 Nuritas Limited Topical cosmetic compositions comprising an oligopeptide against anti-aging of the skin
WO2018102612A1 (en) 2016-12-02 2018-06-07 Juno Therapeutics, Inc. Engineered b cells and related compositions and methods
WO2018106782A1 (en) 2016-12-08 2018-06-14 Case Western Reserve University Methods and compositions for enhancing functional myelin production
WO2018104346A1 (en) 2016-12-05 2018-06-14 Nuritas Limited Compositions comprising peptide wkdeagkplvk
WO2018119060A1 (en) 2016-12-20 2018-06-28 Bristol-Myers Squibb Company Methods for increasing the efficiency of homology directed repair (hdr) in the cellular genome
US10011848B2 (en) 2016-11-09 2018-07-03 City University Of Hong Kong System and method for delivery of substance into mammalian cells
EP3354275A1 (en) 2009-02-04 2018-08-01 Sangamo Therapeutics, Inc. Methods and compositions for treating neuropathies
WO2018170333A1 (en) 2017-03-15 2018-09-20 The Broad Institute, Inc. Novel cas13b orthologues crispr enzymes and systems
WO2018187688A1 (en) 2017-04-07 2018-10-11 Massachusetts Institute Of Technology Methods to spatially profile protease activity in tissue and sections
WO2018191388A1 (en) 2017-04-12 2018-10-18 The Broad Institute, Inc. Novel type vi crispr orthologs and systems
US10117911B2 (en) 2015-05-29 2018-11-06 Agenovir Corporation Compositions and methods to treat herpes simplex virus infections
WO2018208910A1 (en) 2017-05-09 2018-11-15 The Broad Institute Inc. Gut microbiome function predicts response to anti-integrin biologic therapy in inflammatory bowel diseases
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
WO2018229521A1 (en) 2016-06-16 2018-12-20 Oslo Universitetssykehus Hf Improved gene editing
WO2018234370A1 (en) 2017-06-20 2018-12-27 Institut Curie Immune cells defective for suv39h1
WO2019005884A1 (en) 2017-06-26 2019-01-03 The Broad Institute, Inc. Crispr/cas-adenine deaminase based compositions, systems, and methods for targeted nucleic acid editing
WO2019023680A1 (en) 2017-07-28 2019-01-31 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (pace)
US10195280B2 (en) 2014-07-15 2019-02-05 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
WO2019060746A1 (en) 2017-09-21 2019-03-28 The Broad Institute, Inc. Systems, methods, and compositions for targeted nucleic acid editing
WO2019075292A1 (en) 2017-10-12 2019-04-18 Massachusetts Institute Of Technology Prostate cancer protease nanosensors and uses thereof
WO2019089982A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
US10287565B2 (en) 2014-07-28 2019-05-14 Sk Innovation Co., Ltd. Isoprene synthase and method of preparing isoprene using thereof
WO2019100023A1 (en) 2017-11-17 2019-05-23 Iovance Biotherapeutics, Inc. Til expansion from fine needle aspirates and small biopsies
WO2019103857A1 (en) 2017-11-22 2019-05-31 Iovance Biotherapeutics, Inc. Expansion of peripheral blood lymphocytes (pbls) from peripheral blood
EP3492593A1 (en) 2013-11-13 2019-06-05 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
EP3498833A1 (en) 2011-09-21 2019-06-19 Sangamo Therapeutics, Inc. Methods and compositions for regulation of transgene expression
WO2019136456A1 (en) 2018-01-08 2019-07-11 Iovance Biotherapeutics, Inc. Processes for generating til products enriched for tumor antigen-specific t-cells
WO2019136459A1 (en) 2018-01-08 2019-07-11 Iovance Biotherapeutics, Inc. Processes for generating til products enriched for tumor antigen-specific t-cells
WO2019143677A1 (en) 2018-01-17 2019-07-25 Vertex Pharmaceuticals Incorporated Quinoxalinone compounds, compositions, methods, and kits for increasing genome editing efficiency
WO2019143678A1 (en) 2018-01-17 2019-07-25 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
WO2019143675A1 (en) 2018-01-17 2019-07-25 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
WO2019160383A1 (en) 2018-02-19 2019-08-22 고려대학교 산학협력단 Vaccine comprising epitope of heat shock protein, and use thereof
WO2019173332A1 (en) 2018-03-05 2019-09-12 Massachusetts Institute Of Technology Inhalable nanosensors with volatile reporters and uses thereof
WO2019226953A1 (en) 2018-05-23 2019-11-28 The Broad Institute, Inc. Base editors and uses thereof
WO2019236566A1 (en) 2018-06-05 2019-12-12 Lifeedit, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
US10544405B2 (en) 2013-01-16 2020-01-28 Emory University Cas9-nucleic acid complexes and uses related thereto
WO2020033601A1 (en) 2018-08-07 2020-02-13 The Broad Institute, Inc. Novel cas12b enzymes and systems
WO2020041380A1 (en) 2018-08-20 2020-02-27 The Broad Institute, Inc. Methods and compositions for optochemical control of crispr-cas9
GB202000934D0 (en) 2020-01-22 2020-03-04 Ucl Business Ltd Engineered immune cells
WO2020065062A1 (en) 2018-09-28 2020-04-02 Wageningen Universiteit Off-target activity inhibitors for guided endonucleases
WO2020069029A1 (en) 2018-09-26 2020-04-02 Emendobio Inc. Novel crispr nucleases
WO2020068920A2 (en) 2018-09-25 2020-04-02 Massachusetts Institute Of Technology Lung protease nanosensors and uses thereof
WO2020069373A1 (en) 2018-09-28 2020-04-02 President And Fellows Of Harvard College Cellular reprogramming to reverse aging and promote organ and tissue regeneration
WO2020081635A2 (en) 2018-10-16 2020-04-23 Massachusetts Institute Of Technology Renal clearable nanocatalysts for disease monitoring
WO2020079033A1 (en) 2018-10-15 2020-04-23 Fondazione Telethon Genome editing methods and constructs
WO2020092453A1 (en) 2018-10-29 2020-05-07 The Broad Institute, Inc. Nucleobase editors comprising geocas9 and uses thereof
WO2020096988A2 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
WO2020096989A1 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients refractory for anti-pd-1 antibody
WO2020096986A2 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Selection of improved tumor reactive t-cells
WO2020096927A1 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Expansion of tils utilizing akt pathway inhibitors
EP3653229A1 (en) 2013-12-12 2020-05-20 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for genome editing
WO2020102659A1 (en) 2018-11-15 2020-05-22 The Broad Institute, Inc. G-to-t base editors and uses thereof
US10676749B2 (en) 2013-02-07 2020-06-09 The General Hospital Corporation Tale transcriptional activators
WO2020131862A1 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Crispr-associated transposase systems and methods of use thereof
WO2020131547A1 (en) 2018-12-19 2020-06-25 Iovance Biotherapeutics, Inc. Methods of expanding tumor infiltrating lymphocytes using engineered cytokine receptor pairs and uses thereof
EP3674411A1 (en) 2013-06-17 2020-07-01 The Broad Institute, Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
EP3674408A1 (en) 2014-06-16 2020-07-01 The Johns Hopkins University Compositions and methods for the expression of crispr guide rnas
WO2020139783A2 (en) 2018-12-27 2020-07-02 Lifeedit, Inc. Polypeptides useful for gene editing and methods of use
WO2020150560A1 (en) 2019-01-17 2020-07-23 Massachusetts Institute Of Technology Sensors for detecting and imaging of cancer metastasis
US10724020B2 (en) 2016-02-02 2020-07-28 Sangamo Therapeutics, Inc. Compositions for linking DNA-binding domains and cleavage domains
EP3686279A1 (en) 2014-08-17 2020-07-29 The Broad Institute, Inc. Genome editing using cas9 nickases
US10738278B2 (en) 2014-07-15 2020-08-11 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
WO2020163379A1 (en) 2019-02-05 2020-08-13 Emendobio Inc. Crispr compositions and methods for promoting gene editing of ribosomal protein s19 (rps19) gene
WO2020163307A1 (en) 2019-02-06 2020-08-13 Emendobio Inc. New engineered high fidelity cas9
WO2020172343A2 (en) 2019-02-19 2020-08-27 Massachusetts Institute Of Technology Methods for treating injuries
WO2020181180A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. A:t to c:g base editors and uses thereof
WO2020181178A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. T:a to a:t base editing through thymine alkylation
WO2020181202A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. A:t to t:a base editing through adenine deamination and oxidation
WO2020181193A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. T:a to a:t base editing through adenosine methylation
WO2020180733A1 (en) 2019-03-01 2020-09-10 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof
WO2020181195A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. T:a to a:t base editing through adenine excision
WO2020183197A1 (en) 2019-03-14 2020-09-17 Ucl Business Ltd Minimal promoter
WO2020191102A1 (en) 2019-03-18 2020-09-24 The Broad Institute, Inc. Type vii crispr proteins and systems
WO2020191153A2 (en) 2019-03-19 2020-09-24 The Broad Institute, Inc. Methods and compositions for editing nucleotide sequences
US10786533B2 (en) 2015-07-15 2020-09-29 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
WO2020205838A1 (en) 2019-04-02 2020-10-08 Sangamo Therapeutics, Inc. Methods for the treatment of beta-thalassemia
WO2020210751A1 (en) 2019-04-12 2020-10-15 The Broad Institute, Inc. System for genome editing
US10808247B2 (en) 2015-07-06 2020-10-20 Phio Pharmaceuticals Corp. Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
EP3725885A1 (en) 2013-06-17 2020-10-21 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions methods, screens and applications thereof
WO2020214842A1 (en) 2019-04-17 2020-10-22 The Broad Institute, Inc. Adenine base editors with reduced off-target effects
EP3730615A2 (en) 2013-05-15 2020-10-28 Sangamo Therapeutics, Inc. Methods and compositions for treatment of a genetic condition
WO2020223539A1 (en) 2019-04-30 2020-11-05 The Broad Institute, Inc. Methods and compositions for barcoding nucleic acid libraries and cell populations
WO2020232029A1 (en) 2019-05-13 2020-11-19 Iovance Biotherapeutics, Inc. Methods and compositions for selecting tumor infiltrating lymphocytes and uses of the same in immunotherapy
WO2020232271A1 (en) 2019-05-14 2020-11-19 The Broad Institute, Inc. Compositions and methods for targeting multinucleated cells
WO2020236982A1 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Aav delivery of nucleobase editors
WO2020236972A2 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Non-class i multi-component nucleic acid targeting systems
WO2020243276A1 (en) 2019-05-29 2020-12-03 Altria Client Services Llc Compositions and methods for producing tobacco plants and products having reduced or eliminated suckers
WO2020243485A1 (en) 2019-05-29 2020-12-03 Massachusetts Institute Of Technology Hiv-1 specific immunogen compositions and methods of use
WO2020243371A1 (en) 2019-05-28 2020-12-03 Massachusetts Institute Of Technology Methods and compositions for modulating immune responses
EP3747425A1 (en) 2015-07-16 2020-12-09 Nuritas Limited Anti-inflammatory peptides, and uses thereof
EP3757116A1 (en) 2013-12-09 2020-12-30 Sangamo Therapeutics, Inc. Methods and compositions for genome engineering
WO2021001534A1 (en) 2019-07-03 2021-01-07 Wageningen Universiteit Crispr type v-u1 system from mycobacterium mucogenicum and uses thereof
WO2021013950A1 (en) 2019-07-23 2021-01-28 Mnemo Therapeutics Immune cells defective for suv39h1
WO2021025750A1 (en) 2019-08-08 2021-02-11 The Broad Institute, Inc. Base editors with diversified targeting scope
WO2021030666A1 (en) 2019-08-15 2021-02-18 The Broad Institute, Inc. Base editing by transglycosylation
WO2021030344A1 (en) 2019-08-12 2021-02-18 Lifeedit, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2021034717A1 (en) 2019-08-16 2021-02-25 Massachusetts Institute Of Technology Targeted trans-splicing using crispr/cas13
WO2021045292A1 (en) 2019-09-03 2021-03-11 주식회사 제노포커스 Crm197 protein expression method
US10945955B2 (en) 2018-04-25 2021-03-16 University Of Massachusetts Artificial exosome composition and related methods
WO2021072328A1 (en) 2019-10-10 2021-04-15 The Broad Institute, Inc. Methods and compositions for prime editing rna
WO2021072248A2 (en) 2019-10-10 2021-04-15 Altria Client Services Llc Pale yellow locus and its applications in tobacco
WO2021087361A1 (en) 2019-11-01 2021-05-06 Sangamo Therapeutics, Inc. Zinc finger nuclease variants for treating or preventing lysosomal storage diseases
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
WO2021092464A2 (en) 2019-11-08 2021-05-14 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides targeting bromodomain containing protein 4 (brd4) for immunotherapy
EP3825406A1 (en) 2013-06-17 2021-05-26 The Broad Institute Inc. Delivery and use of the crispr-cas systems, vectors and compositions for hepatic targeting and therapy
WO2021108717A2 (en) 2019-11-26 2021-06-03 The Broad Institute, Inc Systems and methods for evaluating cas9-independent off-target editing of nucleic acids
WO2021118990A1 (en) 2019-12-11 2021-06-17 Iovance Biotherapeutics, Inc. Processes for the production of tumor infiltrating lymphocytes (tils) and methods of using the same
WO2021126435A1 (en) 2019-12-20 2021-06-24 Novarock Biotherapeutics, Ltd. Anti-interleukin-23 p19 antibodies and methods of use thereof
WO2021138247A1 (en) 2019-12-30 2021-07-08 LifeEDIT Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2021138537A1 (en) 2019-12-31 2021-07-08 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides with improved systemic delivery
WO2021155065A1 (en) 2020-01-28 2021-08-05 The Broad Institute, Inc. Base editors, compositions, and methods for modifying the mitochondrial genome
WO2021158999A1 (en) 2020-02-05 2021-08-12 The Broad Institute, Inc. Gene editing methods for treating spinal muscular atrophy
WO2021158921A2 (en) 2020-02-05 2021-08-12 The Broad Institute, Inc. Adenine base editors and uses thereof
WO2021163191A1 (en) 2020-02-10 2021-08-19 Board Of Regents, The University Of Texas System Methods for rapid cloning and expression of hla class i cells
WO2021167687A1 (en) 2020-02-18 2021-08-26 Massachusetts Institute Of Technology Multiplexed in vivo disease sensing with nucleic acid-barcoded reporters
US11124796B2 (en) 2014-09-24 2021-09-21 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for modeling competition of multiple cancer mutations in vivo
WO2021217002A1 (en) 2020-04-24 2021-10-28 Lifeedit Therapeutics, Inc . Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2021222318A1 (en) 2020-04-28 2021-11-04 The Broad Institute, Inc. Targeted base editing of the ush2a gene
WO2021226061A1 (en) 2020-05-04 2021-11-11 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
WO2021224416A1 (en) 2020-05-06 2021-11-11 Cellectis S.A. Methods to genetically modify cells for delivery of therapeutic proteins
WO2021224395A1 (en) 2020-05-06 2021-11-11 Cellectis S.A. Methods for targeted insertion of exogenous sequences in cellular genomes
WO2021231437A1 (en) 2020-05-11 2021-11-18 LifeEDIT Therapeutics, Inc. Rna-guided nucleic acid binding proteins and active fragments and variants thereof and methods of use
EP3916086A1 (en) 2017-06-23 2021-12-01 Inscripta, Inc. Nucleic acid-guided nucleases
US11214780B2 (en) 2015-10-23 2022-01-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US11219695B2 (en) 2016-10-20 2022-01-11 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of Fabry disease
EP3939596A1 (en) 2020-07-14 2022-01-19 Bimeda Animal Health Limited A composition for treating helminth infestation in a mammal
WO2022015969A1 (en) 2020-07-15 2022-01-20 LifeEDIT Therapeutics, Inc. Uracil stabilizing proteins and active fragments and variants thereof and methods of use
WO2022023576A1 (en) 2020-07-30 2022-02-03 Institut Curie Immune cells defective for socs1
WO2022034374A2 (en) 2020-08-11 2022-02-17 University Of Oslo Improved gene editing
WO2022046760A2 (en) 2020-08-25 2022-03-03 Kite Pharma, Inc. T cells with improved functionality
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
WO2022051591A2 (en) 2020-09-04 2022-03-10 Novarock Biotherapeutics, Ltd. Nectin-4 antibodies and uses thereof
WO2022056254A2 (en) 2020-09-11 2022-03-17 LifeEDIT Therapeutics, Inc. Dna modifying enzymes and active fragments and variants thereof and methods of use
WO2022060927A1 (en) 2020-09-17 2022-03-24 Northwestern University Engineered mammalian genetic circuits and methods of using the same
US11299755B2 (en) 2013-09-06 2022-04-12 President And Fellows Of Harvard College Switchable CAS9 nucleases and uses thereof
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
WO2022081015A1 (en) 2020-10-16 2022-04-21 Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis Anti-tumor immunity induces the presentation of aberrant peptides
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
WO2022150790A2 (en) 2021-01-11 2022-07-14 The Broad Institute, Inc. Prime editor variants, constructs, and methods for enhancing prime editing efficiency and precision
EP4032538A2 (en) 2009-03-02 2022-07-27 Massachusetts Institute of Technology Methods and products for in vivo enzyme profiling
WO2022164319A1 (en) 2021-01-29 2022-08-04 Wageningen Universiteit Crispr-associated base-editing of the complementary strand
WO2022169836A1 (en) 2021-02-03 2022-08-11 Altria Client Services Llc Terpene production in plants
WO2022192789A1 (en) 2021-03-12 2022-09-15 Synthego Corporation Genetically modified cells expressing antigen-containing fusion proteins and uses thereof
WO2022204093A1 (en) 2021-03-22 2022-09-29 LifeEDIT Therapeutics, Inc. Dna modifyng enzymes and active fragments and variants thereof and methods of use
WO2022219168A1 (en) 2021-04-14 2022-10-20 University College Cork - National University Of Ireland, Cork Psg1 for use in the treatment of osteoarthritis
WO2022219008A1 (en) 2021-04-14 2022-10-20 University College Cork - National University Of Ireland, Cork Treatment of cerebrovascular events and neurological disorders
WO2022243565A1 (en) 2021-05-21 2022-11-24 Cellectis S.A. Enhancing efficacy of t-cell-mediated immunotherapy by modulating cancer-associated fibroblasts in solid tumors
WO2022261394A1 (en) 2021-06-11 2022-12-15 LifeEDIT Therapeutics, Inc. Rna polymerase iii promoters and methods of use
WO2022261509A1 (en) 2021-06-11 2022-12-15 The Broad Institute, Inc. Improved cytosine to guanine base editors
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US11559486B2 (en) 2015-07-22 2023-01-24 Acrotech Biopharma, LLC Ready-to-use formulation for Vincristine Sulfate Liposome Injection
WO2023015264A1 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Immunotherapy of cancer utilizing natural killer cells treated with chemically modified oligonucleotides
WO2023015309A2 (en) 2021-08-06 2023-02-09 The Broad Institute, Inc. Improved prime editors and methods of use
WO2023015265A2 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides
US11578343B2 (en) 2014-07-30 2023-02-14 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US11578334B2 (en) 2017-10-25 2023-02-14 Monsanto Technology Llc Targeted endonuclease activity of the RNA-guided endonuclease CasX in eukaryotes
WO2023055239A1 (en) 2021-10-01 2023-04-06 Koninklijke Nederlandse Akademie Van Wetenschappen Mammalian cardiac regeneration
DE112021002672T5 (en) 2020-05-08 2023-04-13 President And Fellows Of Harvard College METHODS AND COMPOSITIONS FOR EDIT BOTH STRANDS SIMULTANEOUSLY OF A DOUBLE STRANDED NUCLEOTIDE TARGET SEQUENCE
WO2023077148A1 (en) 2021-11-01 2023-05-04 Tome Biosciences, Inc. Single construct platform for simultaneous delivery of gene editing machinery and nucleic acid cargo
WO2023076898A1 (en) 2021-10-25 2023-05-04 The Broad Institute, Inc. Methods and compositions for editing a genome with prime editing and a recombinase
WO2023080788A1 (en) 2021-11-05 2023-05-11 Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis Tryptophan depletion induces production and presentation of tryptophan to phenylalanine substitutions
US11649444B1 (en) 2021-11-02 2023-05-16 Huidagene Therapeutics Co., Ltd. CRISPR-CAS12i systems
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
WO2023096494A1 (en) 2021-11-26 2023-06-01 Synvolux Ip B.V. Conserved coronavirus t cell epitopes
WO2023122764A1 (en) 2021-12-22 2023-06-29 Tome Biosciences, Inc. Co-delivery of a gene editor construct and a donor template
US11692182B2 (en) 2015-10-09 2023-07-04 Monsanto Technology Llc RNA-guided DNA nucleases and uses thereof
WO2023126458A1 (en) 2021-12-28 2023-07-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
US11702651B2 (en) 2016-08-03 2023-07-18 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
WO2023139557A1 (en) 2022-01-24 2023-07-27 LifeEDIT Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
US11713446B2 (en) 2018-01-08 2023-08-01 Iovance Biotherapeutics, Inc. Processes for generating TIL products enriched for tumor antigen-specific T-cells
WO2023150637A1 (en) 2022-02-02 2023-08-10 Inscripta, Inc. Nucleic acid-guided nickase fusion proteins
EP4234570A2 (en) 2018-09-18 2023-08-30 Sangamo Therapeutics, Inc. Programmed cell death 1 (pd1) specific nucleases
EP4245853A2 (en) 2013-06-17 2023-09-20 The Broad Institute, Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
US11766488B2 (en) 2016-07-05 2023-09-26 The Johns Hopkins University Compositions and methods comprising improvements of CRISPR guide RNAS using the H1 promoter
WO2023196802A1 (en) 2022-04-04 2023-10-12 The Broad Institute, Inc. Cas9 variants having non-canonical pam specificities and uses thereof
WO2023196818A1 (en) 2022-04-04 2023-10-12 The Regents Of The University Of California Genetic complementation compositions and methods
WO2023196851A1 (en) 2022-04-06 2023-10-12 President And Fellows Of Harvard College Reversing aging of the central nervous system
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
WO2023205744A1 (en) 2022-04-20 2023-10-26 Tome Biosciences, Inc. Programmable gene insertion compositions
WO2023212715A1 (en) 2022-04-28 2023-11-02 The Broad Institute, Inc. Aav vectors encoding base editors and uses thereof
WO2023215831A1 (en) 2022-05-04 2023-11-09 Tome Biosciences, Inc. Guide rna compositions for programmable gene insertion
WO2023220457A1 (en) 2022-05-13 2023-11-16 Northwestern University Receptor engagement-mediated enhancement of biologics delivery
WO2023220459A1 (en) 2022-05-13 2023-11-16 Northwestern University Active loading of cargo entity into lipid bilayer particles using dimerization domains
WO2023220392A1 (en) 2022-05-13 2023-11-16 Northwestern University Synthetic modular extracellular sensors that employ natural receptor ligand-binding domains
US11820969B2 (en) 2016-12-23 2023-11-21 President And Fellows Of Harvard College Editing of CCR2 receptor gene to protect against HIV infection
WO2023222928A2 (en) 2022-05-20 2023-11-23 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023225670A2 (en) 2022-05-20 2023-11-23 Tome Biosciences, Inc. Ex vivo programmable gene insertion
WO2023230613A1 (en) 2022-05-27 2023-11-30 The Broad Institute, Inc. Improved mitochondrial base editors and methods for editing mitochondrial dna
WO2023230433A1 (en) 2022-05-23 2023-11-30 Altria Client Services Llc Methods and compositions for regulating alkaloids in tobacco field
WO2023240137A1 (en) 2022-06-08 2023-12-14 The Board Institute, Inc. Evolved cas14a1 variants, compositions, and methods of making and using same in genome editing
WO2024003334A1 (en) 2022-06-30 2024-01-04 Cellectis S.A. Enhancing safety of t-cell-mediated immunotherapy
US11866469B2 (en) 2019-02-06 2024-01-09 Klogenix Llc DNA binding proteins and uses thereof
US11872262B2 (en) 2017-05-09 2024-01-16 Vib Vzw Means and methods for treating bacterial infections
WO2024015383A1 (en) 2022-07-12 2024-01-18 Northwestern University Engineered hypoxia biosensors and methods of using the same
WO2024020587A2 (en) 2022-07-22 2024-01-25 Tome Biosciences, Inc. Pleiopluripotent stem cell programmable gene insertion
WO2024031038A1 (en) 2022-08-05 2024-02-08 Altria Client Services Llc Methods and compositions for regulating alkaloids in tobacco
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
WO2024033901A1 (en) 2022-08-12 2024-02-15 LifeEDIT Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2024040083A1 (en) 2022-08-16 2024-02-22 The Broad Institute, Inc. Evolved cytosine deaminases and methods of editing dna using same
WO2024042489A1 (en) 2022-08-25 2024-02-29 LifeEDIT Therapeutics, Inc. Chemical modification of guide rnas with locked nucleic acid for rna guided nuclease-mediated gene editing
US11920181B2 (en) 2013-08-09 2024-03-05 President And Fellows Of Harvard College Nuclease profiling system
WO2024047587A1 (en) 2022-08-31 2024-03-07 Regel Therapeutics, Inc. Cas-phi compositions and methods of use
EP4335925A2 (en) 2019-04-09 2024-03-13 The Regents of the University of California Long-lasting analgesia via targeted in vivo epigenetic repression
US11959094B2 (en) 2023-05-02 2024-04-16 Salk Institute For Biological Studies Methods and compositions for genome editing in non-dividing cells

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4485054A (en) * 1982-10-04 1984-11-27 Lipoderm Pharmaceuticals Limited Method of encapsulating biologically active materials in multilamellar lipid vesicles (MLV)
US4774085A (en) * 1985-07-09 1988-09-27 501 Board of Regents, Univ. of Texas Pharmaceutical administration systems containing a mixture of immunomodulators
US4897355A (en) * 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4485054A (en) * 1982-10-04 1984-11-27 Lipoderm Pharmaceuticals Limited Method of encapsulating biologically active materials in multilamellar lipid vesicles (MLV)
US4897355A (en) * 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4774085A (en) * 1985-07-09 1988-09-27 501 Board of Regents, Univ. of Texas Pharmaceutical administration systems containing a mixture of immunomodulators

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BIOCHEMICA ET BIOPHYSICA ACTA, Volume 858, issued 1986, L.D. MAYER et al., "Vesicles of Variable Sizes Produced by a Rapid Extrusion Procedure", pages 161-168. *
BIOCHEMISTRY, Volume 27, issued 1988, L. STAMATATOS et al., "Interactions of Cationoc Lipid Vessicles with Negatively Charged Phospholipid Vesicles and Biological Membranes", pages 3917-3925. *

Cited By (602)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5667764A (en) * 1988-05-02 1997-09-16 Zynaxis, Inc. Compounds, compositions and methods for binding bio-affecting substances to surface membranes of bio-particles
EP0523189A4 (en) * 1990-04-19 1995-04-26 Vical Inc
US5459127A (en) * 1990-04-19 1995-10-17 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
EP0523189A1 (en) * 1990-04-19 1993-01-20 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5693769A (en) * 1991-12-13 1997-12-02 Transcell Technologies, Inc. Glycosylated steroid derivatives for transport across biological membranes and process for making and using same
US5795870A (en) * 1991-12-13 1998-08-18 Trustees Of Princeton University Compositions and methods for cell transformation
US5770580A (en) * 1992-04-13 1998-06-23 Baylor College Of Medicine Somatic gene therapy to cells associated with fluid spaces
US6890554B2 (en) 1993-06-01 2005-05-10 Invitrogen Corporation Genetic immunization with cationic lipids
US7166298B2 (en) 1993-06-01 2007-01-23 Invitrogen Corporation Genetic immunization with cationic lipids
US7501542B2 (en) 1993-12-20 2009-03-10 Invitrogen Corporation Highly-packed polycationic ammonium, sulfonium and phosphonium lipids
US6716882B2 (en) 1993-12-20 2004-04-06 Invitrogen Corporation Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US6136792A (en) * 1994-01-13 2000-10-24 Calydon, Inc. Prostate specific enhancer polynucleotides and methods of use thereof
US6057299A (en) * 1994-01-13 2000-05-02 Calydon, Inc. Tissue-specific enhancer active in prostate
US5928944A (en) * 1994-02-04 1999-07-27 The United States Of America As Represented By The Department Of Health And Human Services Method of adenoviral-medicated cell transfection
US7687070B2 (en) 1994-02-11 2010-03-30 Life Technologies Corporation Reagents for intracellular delivery of macromolecules
US6989434B1 (en) 1994-02-11 2006-01-24 Invitrogen Corporation Reagents for intracellular delivery of macromolecules
US6773719B2 (en) 1994-03-04 2004-08-10 Esperion Luv Development, Inc. Liposomal compositions, and methods of using liposomal compositions to treat dislipidemias
US7101570B2 (en) 1994-03-04 2006-09-05 The University Of British Columbia Liposome compositions and methods for the treatment of atherosclerosis
US6312719B1 (en) 1994-03-04 2001-11-06 The University Of British Columbia Liposome compositions and methods for the treatment of atherosclerosis
US6995008B1 (en) 1994-03-07 2006-02-07 Merck & Co., Inc. Coordinate in vivo gene expression
US7235236B2 (en) 1995-02-28 2007-06-26 The Regents Of The University Of California Polynucleotide encoding human adenylylcyclase VI and uses thereof for enhancing cardiac function
US6752987B1 (en) 1995-02-28 2004-06-22 The Regents Of The University Of California Adenovirus encoding human adenylylcyclase (AC) VI
US6174871B1 (en) 1995-02-28 2001-01-16 The Regents Of The University Of California Gene therapies for enhancing cardiac function
US6139871A (en) * 1995-07-26 2000-10-31 The University Of British Columbia Liposome compositions and methods for the treatment of atherosclerosis
US6367479B1 (en) 1995-10-11 2002-04-09 Esperion Luv Development, Inc. Method of forcing the reverse transport of cholesterol from a body part to the liver while avoiding harmful disruptions of hepatic cholesterol homeostasis, and pharmaceutical compositions and kit related thereto
US6670332B1 (en) 1995-11-30 2003-12-30 Vical Incorporated Complex cationic lipids having quarternary nitrogens therein
WO1997019675A2 (en) * 1995-11-30 1997-06-05 Vical Incorporated Complex cationic lipids
JP2008247906A (en) * 1995-11-30 2008-10-16 Vical Inc Cationic lipid complex
WO1997019675A3 (en) * 1995-11-30 1997-10-02 Vical Inc Complex cationic lipids
US8541628B2 (en) 1995-11-30 2013-09-24 Vical Incorporated Complex cationic lipids having quaternary nitrogens therein
US7790696B2 (en) 1996-09-13 2010-09-07 Lipoxen Technologies Limited Cationic liposomes containing immune response generating moieties
EP1254657A3 (en) * 1996-09-13 2003-03-05 Lipoxen Technologies Limited Liposomes
JP2001502299A (en) * 1996-09-13 2001-02-20 リポゼン リミテッド Liposome
WO1998010748A1 (en) * 1996-09-13 1998-03-19 The School Of Pharmacy Liposomes
AU728581B2 (en) * 1996-09-13 2001-01-11 Lipoxen Limited Liposomes
EP1254657A2 (en) * 1996-09-13 2002-11-06 Lipoxen Technologies Limited Liposomes
EP1783139A2 (en) 1996-10-01 2007-05-09 Geron Corporation Human telomerase catalytic subunit
EP1333094A2 (en) 1996-10-01 2003-08-06 Geron Corporation Human telomerase catalytic subunit
EP2213740A1 (en) 1996-10-01 2010-08-04 The Regents of the University of Colorado Human telomerase catalytic subunit
WO1998014593A2 (en) 1996-10-01 1998-04-09 Geron Corporation Human telomerase catalytic subunit
WO1999006026A1 (en) * 1997-07-30 1999-02-11 Biovector Therapeutics (S.A.) Stable particulate complexes with neutral or negative global charge of lamellar structure
US6096335A (en) * 1997-07-30 2000-08-01 Biovector Therapeutics, S.A. Stable particulate complexes having a lamellar, rolled, and condensed structure
FR2766705A1 (en) * 1997-07-30 1999-02-05 Biovector Therapeutics Stable particulate complexes for use in gene therapy
FR2766706A1 (en) * 1997-07-30 1999-02-05 Biovector Therapeutics Sa STABLE, NEUTRAL OR NEGATIVE GLOBAL PARTICLE COMPLEXES OF A MULTILAMELLAR STRUCTURE COMPRISING AT LEAST ONE GLOBALLY ANIONIC BIOLOGICALLY ACTIVE SUBSTANCE AND A CATIONIC COMPONENT, THEIR PREPARATION AND USE
US6916918B2 (en) 1997-08-04 2005-07-12 Cell Genesys, Inc. Human glandular kallikrein enhancer, vectors comprising the enhancer and methods of use thereof
US7462605B2 (en) 1998-01-23 2008-12-09 Celmed Oncology (Usa), Inc. Phosphoramidate compounds and methods of use
US7601703B2 (en) 1998-01-23 2009-10-13 Celmed Oncology (Usa), Inc. Enzyme catalyzed therapeutic agents
US6245750B1 (en) 1998-01-23 2001-06-12 Newbiotics, Inc. Enzyme catalyzed therapeutic agents
US6339151B1 (en) 1998-01-23 2002-01-15 Newbiotics, Inc. Enzyme catalyzed therapeutic agents
US7414026B2 (en) 1998-05-06 2008-08-19 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Inhibitors of NF-kappaB activation
US9358300B2 (en) 1998-11-12 2016-06-07 Life Technologies Corporation Transfection reagents
US8435960B2 (en) 1998-12-30 2013-05-07 Lakewood-Amedex, Inc. Devices for improved wound management
US7868162B2 (en) 1998-12-30 2011-01-11 Lakewood-Amedex, Inc. Antimicrobial and antiviral compounds and methods for their use
EP2397549A2 (en) 1999-02-04 2011-12-21 BP Corporation North America Inc. Non-stochastic generation of genetic vaccines and enzymes
US7887836B2 (en) 1999-04-01 2011-02-15 Hana Biosciences, Inc. Compositions and methods for treating lymphoma
EP1985285A2 (en) 1999-04-01 2008-10-29 Hana Biosciences, Inc. Compositions and methods for treating lymphoma
US7247316B2 (en) 1999-04-01 2007-07-24 Board Of Regents, The University Of Texas System Compositions and methods for treating lymphoma
US7311924B2 (en) 1999-04-01 2007-12-25 Hana Biosciences, Inc. Compositions and methods for treating cancer
EP2266537A2 (en) 1999-04-01 2010-12-29 Hana Biosciences, Inc. Compositions and methods for treating cancer
US7244450B2 (en) 1999-04-01 2007-07-17 Inex Pharmaceuticals Corporation Compositions and methods for treating lymphoma
WO2000063366A2 (en) * 1999-04-20 2000-10-26 Sequitur, Inc. Improved antisense oligomers
WO2000063366A3 (en) * 1999-04-20 2001-01-18 Sequitur Inc Improved antisense oligomers
US7384923B2 (en) 1999-05-14 2008-06-10 Lipoxen Technologies Limited Liposomes
US7419968B1 (en) 1999-07-22 2008-09-02 Celmed Oncology (Usa), Inc. Methods for treating therapy-resistant tumors
US6683061B1 (en) 1999-07-22 2004-01-27 Newbiotics, Inc. Enzyme catalyzed therapeutic activation
US7605144B2 (en) 1999-07-22 2009-10-20 Celmed Oncology (Usa), Inc. Enzyme catalyzed therapeutic compounds
EP2207032A1 (en) 2000-02-08 2010-07-14 Sangamo BioSciences, Inc. Cells expressing zinc finger protein for drug discovery
US7138388B2 (en) 2001-01-19 2006-11-21 Celmed Oncology (Usa), Inc. Methods to treat autoimmune and inflammatory conditions
EP2311466A2 (en) 2001-05-03 2011-04-20 Lakewood-Amedex, Inc Antimicrobial compounds and methods for their use
US7094756B2 (en) 2001-06-22 2006-08-22 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Abin-mediated hepatitis protection
US8916529B2 (en) 2001-07-10 2014-12-23 Lakewood-Amedex, Inc. Oligonucleotide-containing pharmacological compositions and their use
US9567584B2 (en) 2001-07-10 2017-02-14 Lakewood Amedex, Inc. Oligonucleotide—containing pharmacological compositions and their use
US8188259B2 (en) 2001-07-10 2012-05-29 Lakewood-Amedex, Inc. Oligonucleotide-containing pharmacological compositions and their use
US8183361B2 (en) 2001-07-10 2012-05-22 Lakewood-Amedex, Inc. Oligonucleotide-containing pharmacological compositions and their use
EP2351765A2 (en) 2001-07-10 2011-08-03 Lakewood-Amedex, Inc Oligonucleotide-containing pharmacological compositions and their use
US9624509B2 (en) 2001-09-24 2017-04-18 Sangamo Biosciences, Inc. Modulation of stem cells using zinc finger proteins
US8735153B2 (en) 2001-09-24 2014-05-27 Sangamo Biosciences, Inc. Modulation of stem cells using zinc finger proteins
US10888589B2 (en) 2001-09-24 2021-01-12 Sangamo Therapeutics, Inc. Modulation of stem cells using zinc finger proteins
EP2266396A2 (en) 2001-09-24 2010-12-29 Sangamo BioSciences, Inc. Modulation of stem cells using zinc finger proteins
US7122656B2 (en) 2002-01-10 2006-10-17 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Splice variant of MyD88 and uses thereof
US10626398B2 (en) 2002-02-01 2020-04-21 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9796978B1 (en) 2002-02-01 2017-10-24 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US10036025B2 (en) 2002-02-01 2018-07-31 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
EP2128248A1 (en) 2002-02-01 2009-12-02 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
EP2213292A1 (en) 2002-02-01 2010-08-04 Life Technologies Corporation Double-stranded oligonucleotides
US10196640B1 (en) 2002-02-01 2019-02-05 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
EP2924116A1 (en) 2002-02-01 2015-09-30 Life Technologies Corporation Double-stranded oligonucleotides
US10106793B2 (en) 2002-02-01 2018-10-23 Life Technologies Corporation Double-stranded oligonucleotides
US9777275B2 (en) 2002-02-01 2017-10-03 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
EP2455467A1 (en) 2002-02-01 2012-05-23 Life Technologies Corporation Double-stranded oligonucleotides
US9592250B2 (en) 2002-02-01 2017-03-14 Life Technologies Corporation Double-stranded oligonucleotides
EP3415625A1 (en) 2002-02-01 2018-12-19 Life Technologies Corporation Double-stranded oligonucleotides
EP2351836A1 (en) 2002-02-01 2011-08-03 Life Technologies Corporation Double-stranded oligonucleotides
EP2213737A1 (en) 2002-02-01 2010-08-04 Life Technologies Corporation Double-stranded oligonucleotides
US7273620B1 (en) 2002-05-20 2007-09-25 University Of British Columbia Triggered release of liposomal drugs following mixing of cationic and anionic liposomes
US7135324B2 (en) 2002-09-04 2006-11-14 The University Of Connecticut Viral recombinases, related articles, and methods of use thereof
DE10322123A1 (en) * 2003-05-12 2004-12-16 Novosom Ag Stable anionic liposome depot system for prolonged release of protein or peptide drugs, e.g. insulin, comprising phosphatidyl choline, cholesterol, anionic lipid(s) and cationic polymer
EP3222715A1 (en) 2003-08-08 2017-09-27 Sangamo BioSciences, Inc. Methods and compositions for targeted cleavage and recombination
WO2005014791A2 (en) 2003-08-08 2005-02-17 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
US9260726B2 (en) 2003-08-08 2016-02-16 Sangamo Biosciences, Inc. Targeted integration and expression on exogenous nucleic acid sequences
EP2927318A1 (en) 2003-08-08 2015-10-07 Sangamo BioSciences, Inc. Methods and compositions for targeted cleavage and recombination
US7407776B2 (en) 2003-09-19 2008-08-05 Sangamo Biosciences, Inc. Engineered zinc finger proteins for regulation of gene expression
EP2947146A1 (en) 2004-02-05 2015-11-25 Sangamo BioSciences, Inc. Methods and compositions for targeted cleavage and recombination
US7534775B2 (en) 2004-04-08 2009-05-19 Sangamo Biosciences, Inc. Methods and compositions for modulating cardiac contractility
US7811602B2 (en) 2004-05-17 2010-10-12 Tekmira Pharmaceuticals Corporation Liposomal formulations comprising dihydrosphingomyelin and methods of use thereof
US8034598B2 (en) 2004-08-06 2011-10-11 Sangamo Biosciences, Inc. Engineered cleavage half-domains
US8771986B2 (en) 2004-08-06 2014-07-08 Sangamo Biosciences, Inc. Engineered cleavage half-domains
EP2292274A1 (en) 2004-09-16 2011-03-09 Sangamo BioSciences, Inc. Compositions and methods for protein production
US9315862B2 (en) 2004-10-05 2016-04-19 California Institute Of Technology Aptamer regulated nucleic acids and uses thereof
US9309568B2 (en) 2004-10-05 2016-04-12 California Institute Of Technology Aptamer regulated nucleic acids and uses thereof
US7732196B2 (en) 2005-02-28 2010-06-08 Sangamo Biosciences, Inc. Anti-angiogenic methods and compositions
EP2314614A1 (en) 2005-02-28 2011-04-27 Sangamo BioSciences, Inc. Anti-angiogenic methods and compositions
US7795209B2 (en) 2005-02-28 2010-09-14 Sangamo Biosciences, Inc. Anti-angiogenic methods and compositions
US7893022B2 (en) 2005-02-28 2011-02-22 Sangamo Biosciences, Inc. Anti-angiogenic methods and compositions
US8012946B1 (en) 2005-02-28 2011-09-06 Sangamo Biosciences, Inc. Anti-angiogenic methods and compositions
US7358085B2 (en) 2005-02-28 2008-04-15 Sangamo Biosciences, Inc. Anti-angiogenic methods and compositions
WO2006130974A1 (en) 2005-06-08 2006-12-14 Cangene Corporation Hyaluronic acid binding peptides enhance host defense against pathogenic bacteria
US8313925B2 (en) 2005-07-26 2012-11-20 Sangamo Biosciences, Inc. Zinc finger proteins and method for inactivating a dhfr gene in a chinese hamster ovary cell
EP2980220A1 (en) 2005-09-20 2016-02-03 BASF Plant Science GmbH Improved methods controlling gene expression
EP3061834A1 (en) 2005-10-21 2016-08-31 The Regents of the University of California Shp-2 gene mutations in melanoma
WO2007062399A2 (en) 2005-11-23 2007-05-31 The Board Of Regents Of The University Of Texas System Oncogenic ras-specific cytotoxic compound and methods of use thereof
EP2345417A1 (en) 2005-12-22 2011-07-20 Vib Vzw Means and methods for mediating protein interference
US10336791B2 (en) 2005-12-22 2019-07-02 Vib Vzw Means and methods for mediating protein interference
US9095556B2 (en) 2005-12-22 2015-08-04 Vib Vzw Method for inducing protein aggregation using a polypeptide with an aggregation region
US8669418B2 (en) 2005-12-22 2014-03-11 Vib Vzw Means and methods for mediating protein interference
EP2213731A1 (en) 2006-05-25 2010-08-04 Sangamo BioSciences, Inc. Variant foki cleavage half-domains
US7914796B2 (en) 2006-05-25 2011-03-29 Sangamo Biosciences, Inc. Engineered cleavage half-domains
US8158595B2 (en) 2006-11-09 2012-04-17 California Institute Of Technology Modular aptamer-regulated ribozymes
EP2527435A2 (en) 2007-07-12 2012-11-28 Sangamo BioSciences, Inc. Methods and compositions for inactivating alpha 1,6 fucosyltransferase (FUT 8) gene expression
US9040495B2 (en) 2007-08-28 2015-05-26 California Institute Of Technology General composition framework for ligand-controlled RNA regulatory systems
US8367815B2 (en) 2007-08-28 2013-02-05 California Institute Of Technology Modular polynucleotides for ligand-controlled regulatory systems
US8865667B2 (en) 2007-09-12 2014-10-21 California Institute Of Technology Higher-order cellular information processing devices
US9029524B2 (en) 2007-12-10 2015-05-12 California Institute Of Technology Signal activated RNA interference
EP2527439A2 (en) 2007-12-11 2012-11-28 The Scripps Research Institute Compositions and methods related to mRNA translational enhancer elements
EP2610340A1 (en) 2007-12-11 2013-07-03 The Scripps Research Institute Compositions and methods related to mRNA translational enhancer elements
EP2610341A1 (en) 2007-12-11 2013-07-03 The Scripps Research Institute Compositions and methods related to mRNA translational enhancer elements
EP2584038A1 (en) 2007-12-11 2013-04-24 The Scripps Research Institute Compositions and methods related to mRNA translational enhancer elements
US8785611B2 (en) 2007-12-11 2014-07-22 The Scripps Research Institute Compositions and methods related to mRNA translational enhancer elements
EP3643782A1 (en) 2008-02-11 2020-04-29 Phio Pharmaceuticals Corp. Modified rnai polynucleotides and uses thereof
US10633654B2 (en) 2008-02-11 2020-04-28 Phio Pharmaceuticals Corp. Modified RNAi polynucleotides and uses thereof
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
WO2009151591A2 (en) 2008-06-10 2009-12-17 Sangamo Biosciences, Inc. Methods and compositions for generation of bax- and bak-deficient cell lines
EP2910568A2 (en) 2008-06-10 2015-08-26 Sangamo BioSciences, Inc. Methods and compositions for generation of BAX- and BAK-deficient cell lines
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
WO2010021692A1 (en) 2008-08-22 2010-02-25 Sangamo Biosciences, Inc. Methods and compositions for targeted single-stranded cleavage and targeted integration
EP2789691A1 (en) 2008-08-22 2014-10-15 Sangamo BioSciences, Inc. Methods and compositions for targeted single-stranded cleavage and targeted integration
US10041073B2 (en) 2008-09-22 2018-08-07 Rxi Pharmaceuticals Corporation Reduced size self-delivering RNAi compounds
US8664189B2 (en) 2008-09-22 2014-03-04 Rxi Pharmaceuticals Corporation RNA interference in skin indications
US10774330B2 (en) 2008-09-22 2020-09-15 Phio Pharmaceuticals Corp. Reduced size self-delivering RNAI compounds
EP2949752A2 (en) 2008-09-22 2015-12-02 RXi Pharmaceuticals Corporation Reduced size self-delivering rnai compounds
US8796443B2 (en) 2008-09-22 2014-08-05 Rxi Pharmaceuticals Corporation Reduced size self-delivering RNAi compounds
US11396654B2 (en) 2008-09-22 2022-07-26 Phio Pharmaceuticals Corp. Neutral nanotransporters
US9175289B2 (en) 2008-09-22 2015-11-03 Rxi Pharmaceuticals Corporation Reduced size self-delivering RNAi compounds
US10138485B2 (en) 2008-09-22 2018-11-27 Rxi Pharmaceuticals Corporation Neutral nanotransporters
US9303259B2 (en) 2008-09-22 2016-04-05 Rxi Pharmaceuticals Corporation RNA interference in skin indications
EP3336188A1 (en) 2008-09-22 2018-06-20 RXi Pharmaceuticals Corporation Reduced size self-delivering rnai compounds
US10876119B2 (en) 2008-09-22 2020-12-29 Phio Pharmaceuticals Corp. Reduced size self-delivering RNAI compounds
US11254940B2 (en) 2008-11-19 2022-02-22 Phio Pharmaceuticals Corp. Inhibition of MAP4K4 through RNAi
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
EP3156494A1 (en) 2008-12-04 2017-04-19 Sangamo BioSciences, Inc. Genome editing in rats using zinc-finger nucleases
WO2010065123A1 (en) 2008-12-04 2010-06-10 Sangamo Biosciences, Inc. Genome editing in rats using zinc-finger nucleases
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
US10167471B2 (en) 2009-01-05 2019-01-01 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAI
US11667915B2 (en) 2009-02-04 2023-06-06 Phio Pharmaceuticals Corp. RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
EP3354275A1 (en) 2009-02-04 2018-08-01 Sangamo Therapeutics, Inc. Methods and compositions for treating neuropathies
US10479992B2 (en) 2009-02-04 2019-11-19 Phio Pharmaceuticals Corp. RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US8329882B2 (en) 2009-02-18 2012-12-11 California Institute Of Technology Genetic control of mammalian cells with synthetic RNA regulatory systems
EP4032538A2 (en) 2009-03-02 2022-07-27 Massachusetts Institute of Technology Methods and products for in vivo enzyme profiling
US9145555B2 (en) 2009-04-02 2015-09-29 California Institute Of Technology Integrated—ligand-responsive microRNAs
WO2010117464A1 (en) 2009-04-09 2010-10-14 Sangamo Biosciences, Inc. Targeted integration into stem cells
WO2010148010A1 (en) 2009-06-15 2010-12-23 4S3 Bioscience Inc. Methods and compositions for treatment of myotubular myopathy using chimeric polypeptides comprising myotubularih 1 (mtm1) polypeptides
EP2727600A1 (en) 2009-07-28 2014-05-07 Sangamo BioSciences, Inc. Methods and compositions for treating trinucleotide repeat disorders
US9074224B2 (en) 2009-08-03 2015-07-07 Recombinetics, Inc. Methods and compositions for targeted gene modification
WO2011043980A1 (en) 2009-10-07 2011-04-14 Sanford Burnham Medical Research Institute Methods and compositions related to clot-binding lipid compounds
WO2011090804A1 (en) 2010-01-22 2011-07-28 Dow Agrosciences Llc Targeted genomic alteration
EP4328304A2 (en) 2010-02-08 2024-02-28 Sangamo Therapeutics, Inc. Engineered cleavage half-domains
EP2615106A1 (en) 2010-02-08 2013-07-17 Sangamo BioSciences, Inc. Engineered cleavage half-domains
EP3594333A1 (en) 2010-02-08 2020-01-15 Sangamo Therapeutics, Inc. Engineered cleavage half-domains
EP2660318A1 (en) 2010-02-09 2013-11-06 Sangamo BioSciences, Inc. Targeted genomic modification with partially single-stranded donor molecules
WO2011100058A1 (en) 2010-02-09 2011-08-18 Sangamo Biosciences, Inc. Targeted genomic modification with partially single-stranded donor molecules
EP3560503A1 (en) 2010-03-24 2019-10-30 Phio Pharmaceuticals Corp. Rna interference in dermal and fibrotic indications
US10913948B2 (en) 2010-03-24 2021-02-09 Phio Pharmaceuticals Corp. RNA interference in dermal and fibrotic indications
US9963702B2 (en) 2010-03-24 2018-05-08 Rxi Pharmaceuticals Corporation RNA interference in dermal and fibrotic indications
US9340786B2 (en) 2010-03-24 2016-05-17 Rxi Pharmaceuticals Corporation RNA interference in dermal and fibrotic indications
WO2011119887A1 (en) 2010-03-24 2011-09-29 Rxi Pharmaceuticals Corporation Rna interference in dermal and fibrotic indications
WO2011119871A1 (en) 2010-03-24 2011-09-29 Rxi Phrmaceuticals Corporation Rna interference in ocular indications
EP3578183A1 (en) 2010-03-24 2019-12-11 Phio Pharmaceuticals Corp. Rna interference in ocular indications
WO2011130729A2 (en) 2010-04-16 2011-10-20 Salk Institute For Biological Studies Methods for treating metabolic disorders using fgf
EP3156062A1 (en) 2010-05-17 2017-04-19 Sangamo BioSciences, Inc. Novel dna-binding proteins and uses thereof
US9057057B2 (en) 2010-07-27 2015-06-16 The Johns Hopkins University Obligate heterodimer variants of foki cleavage domain
US8956646B2 (en) 2010-08-14 2015-02-17 The Regents Of The University Of California Zwitterionic lipids
WO2012047598A1 (en) 2010-09-27 2012-04-12 Sangamo Biosciences, Inc. Methods and compositions for inhibiting viral entry into cells
EP3511420A1 (en) 2010-09-27 2019-07-17 Sangamo Therapeutics, Inc. Methods and compositions for inhibiting viral entry into cells
WO2012061113A2 (en) 2010-10-25 2012-05-10 Academia Sinica, Taiwan Cancer-targeting peptides and uses thereof in cancer treatment and diagnosis
EP3311822A1 (en) 2010-11-17 2018-04-25 Sangamo Therapeutics, Inc. Methods and compositions for modulating pd1
US10273271B2 (en) 2011-07-15 2019-04-30 The General Hospital Corporation Methods of transcription activator like effector assembly
EP3461896A2 (en) 2011-07-15 2019-04-03 The General Hospital Corporation Methods of transcription activator like effector assembly
US11472849B2 (en) 2011-07-15 2022-10-18 The General Hospital Corporation Methods of transcription activator like effector assembly
WO2013012674A1 (en) 2011-07-15 2013-01-24 The General Hospital Corporation Methods of transcription activator like effector assembly
EP3498833A1 (en) 2011-09-21 2019-06-19 Sangamo Therapeutics, Inc. Methods and compositions for regulation of transgene expression
WO2013130824A1 (en) 2012-02-29 2013-09-06 Sangamo Biosciences, Inc. Methods and compositions for treating huntington's disease
US10894950B2 (en) 2012-05-29 2021-01-19 The General Hospital Corporation TAL-Tet1 fusion proteins and methods of use thereof
EP3747999A1 (en) 2012-05-29 2020-12-09 The General Hospital Corporation Dna modifying fusion proteins and methods of use thereof
EP3744835A1 (en) 2012-05-29 2020-12-02 The General Hospital Corporation Dna modifying fusion proteins and methods of use thereof
US9890364B2 (en) 2012-05-29 2018-02-13 The General Hospital Corporation TAL-Tet1 fusion proteins and methods of use thereof
EP3444342A1 (en) 2012-07-11 2019-02-20 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of lysosomal storage diseases
EP3816281A1 (en) 2012-07-11 2021-05-05 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of lysosomal storage diseases
EP3196301A1 (en) 2012-07-11 2017-07-26 Sangamo BioSciences, Inc. Methods and compositions for the treatment of monogenic diseases
WO2014011237A1 (en) 2012-07-11 2014-01-16 Sangamo Biosciences, Inc. Methods and compositions for the treatment of lysosomal storage diseases
WO2014011901A2 (en) 2012-07-11 2014-01-16 Sangamo Biosciences, Inc. Methods and compositions for delivery of biologics
WO2014036219A2 (en) 2012-08-29 2014-03-06 Sangamo Biosciences, Inc. Methods and compositions for treatment of a genetic condition
WO2014059173A2 (en) 2012-10-10 2014-04-17 Sangamo Biosciences, Inc. T cell modifying compounds and uses thereof
EP3763810A2 (en) 2012-10-10 2021-01-13 Sangamo Therapeutics, Inc. T cell modifying compounds and uses thereof
EP3483185A1 (en) 2012-10-12 2019-05-15 The General Hospital Corporation Transcription activator-like effector (tale) - lysine-specific demethylase 1 (lsd1) fusion proteins
WO2014059255A1 (en) 2012-10-12 2014-04-17 The General Hospital Corporation Transcription activator-like effector (tale) - lysine-specific demethylase 1 (lsd1) fusion proteins
US11891631B2 (en) 2012-10-12 2024-02-06 The General Hospital Corporation Transcription activator-like effector (tale) - lysine-specific demethylase 1 (LSD1) fusion proteins
EP3789405A1 (en) 2012-10-12 2021-03-10 The General Hospital Corporation Transcription activator-like effector (tale) - lysine-specific demethylase 1 (lsd1) fusion proteins
EP3470061A1 (en) 2012-11-20 2019-04-17 Spectrum Pharmaceuticals, Inc. Improved method for the preparation of a dosage of liposome encapsulated vincristine for therapeutic use
US9801874B2 (en) 2012-11-20 2017-10-31 Spectrum Pharmaceuticals Method for the preparation of liposome encapsulated vincristine for therapeutic use
WO2014089541A2 (en) 2012-12-07 2014-06-12 Haplomics, Inc. Factor viii mutation repair and tolerance induction
EP2784162A1 (en) 2012-12-12 2014-10-01 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP2998400A1 (en) 2012-12-12 2016-03-23 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
EP2848690A1 (en) 2012-12-12 2015-03-18 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
WO2014093622A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
EP3825401A1 (en) 2012-12-12 2021-05-26 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
WO2014093635A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
EP2896697A1 (en) 2012-12-12 2015-07-22 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2014093661A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
WO2014093694A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
EP3064585A1 (en) 2012-12-12 2016-09-07 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
WO2014093595A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
EP3144390A1 (en) 2012-12-12 2017-03-22 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP3045537A1 (en) 2012-12-12 2016-07-20 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
EP4234696A2 (en) 2012-12-12 2023-08-30 The Broad Institute Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
EP2840140A1 (en) 2012-12-12 2015-02-25 The Broad Institute, Inc. Crispr-Cas component systems, methods and compositions for sequence manipulation
WO2014093655A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
EP4279588A2 (en) 2012-12-12 2023-11-22 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP4286403A2 (en) 2012-12-12 2023-12-06 The Broad Institute Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
EP4286404A2 (en) 2012-12-12 2023-12-06 The Broad Institute Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
EP2921557A1 (en) 2012-12-12 2015-09-23 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP2940140A1 (en) 2012-12-12 2015-11-04 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP4286402A2 (en) 2012-12-12 2023-12-06 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
EP4299741A2 (en) 2012-12-12 2024-01-03 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
EP3327127A1 (en) 2012-12-12 2018-05-30 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
EP3252160A1 (en) 2012-12-12 2017-12-06 The Broad Institute Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
WO2014093712A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP3705490A1 (en) 2012-12-12 2020-09-09 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
EP3702463A1 (en) 2012-12-12 2020-09-02 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2014093701A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
US11312945B2 (en) 2013-01-16 2022-04-26 Emory University CAS9-nucleic acid complexes and uses related thereto
US10544405B2 (en) 2013-01-16 2020-01-28 Emory University Cas9-nucleic acid complexes and uses related thereto
US10676749B2 (en) 2013-02-07 2020-06-09 The General Hospital Corporation Tale transcriptional activators
US10731167B2 (en) 2013-02-07 2020-08-04 The General Hospital Corporation Tale transcriptional activators
WO2014130723A1 (en) 2013-02-20 2014-08-28 Valerion Therapeutics, Llc Methods and compositions for treatment of pompe disease
US10017581B2 (en) 2013-02-20 2018-07-10 Valerion Therapeutics, Llc Methods and compositions for treatment of Pompe disease
EP3730615A2 (en) 2013-05-15 2020-10-28 Sangamo Therapeutics, Inc. Methods and compositions for treatment of a genetic condition
WO2014190181A1 (en) 2013-05-22 2014-11-27 Northwestern University Rna-directed dna cleavage and gene editing by cas9 enzyme from neisseria meningitidis
EP3778899A1 (en) 2013-05-22 2021-02-17 Northwestern University Rna-directed dna cleavage and gene editing by cas9 enzyme from neisseria meningitidis
WO2014197840A1 (en) 2013-06-07 2014-12-11 Massachusetts Institute Of Technology Affinity-based detection of ligand-encoded synthetic biomarkers
EP3597755A1 (en) 2013-06-17 2020-01-22 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using viral components
WO2014204728A1 (en) 2013-06-17 2014-12-24 The Broad Institute Inc. Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of post mitotic cells
EP3725885A1 (en) 2013-06-17 2020-10-21 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions methods, screens and applications thereof
WO2014204729A1 (en) 2013-06-17 2014-12-24 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using viral components
EP4245853A2 (en) 2013-06-17 2023-09-20 The Broad Institute, Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
EP3674411A1 (en) 2013-06-17 2020-07-01 The Broad Institute, Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
EP3825406A1 (en) 2013-06-17 2021-05-26 The Broad Institute Inc. Delivery and use of the crispr-cas systems, vectors and compositions for hepatic targeting and therapy
US11920181B2 (en) 2013-08-09 2024-03-05 President And Fellows Of Harvard College Nuclease profiling system
WO2015031619A1 (en) 2013-08-28 2015-03-05 Sangamo Biosciences, Inc. Compositions for linking dna-binding domains and cleavage domains
EP3988654A1 (en) 2013-08-28 2022-04-27 Sangamo Therapeutics, Inc. Compositions for linking dna-binding domains and cleavage domains
EP3591045A1 (en) 2013-08-28 2020-01-08 Sangamo Therapeutics, Inc. Compositions for linking dna-binding domains and cleavage domains
US11299755B2 (en) 2013-09-06 2022-04-12 President And Fellows Of Harvard College Switchable CAS9 nucleases and uses thereof
WO2015057980A1 (en) 2013-10-17 2015-04-23 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
EP3441468A2 (en) 2013-10-17 2019-02-13 Sangamo Therapeutics, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
WO2015070212A1 (en) 2013-11-11 2015-05-14 Sangamo Biosciences, Inc. Methods and compositions for treating huntington's disease
EP3492593A1 (en) 2013-11-13 2019-06-05 Children's Medical Center Corporation Nuclease-mediated regulation of gene expression
WO2015085113A1 (en) 2013-12-04 2015-06-11 Rxi Pharmaceuticals Corporation Methods for treatment of wound healing utilizing chemically modified oligonucleotides
EP3757116A1 (en) 2013-12-09 2020-12-30 Sangamo Therapeutics, Inc. Methods and compositions for genome engineering
EP3653229A1 (en) 2013-12-12 2020-05-20 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for genome editing
EP3540051A1 (en) 2013-12-12 2019-09-18 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for hbv and viral diseases and disorders
WO2015089486A2 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Systems, methods and compositions for sequence manipulation with optimized functional crispr-cas systems
EP3470089A1 (en) 2013-12-12 2019-04-17 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components
WO2015089419A2 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components
WO2015089465A1 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for hbv and viral diseases and disorders
EP4183876A1 (en) 2013-12-12 2023-05-24 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for hbv and viral diseases and disorders
WO2015117081A2 (en) 2014-02-03 2015-08-06 Sangamo Biosciences, Inc. Methods and compositions for treatment of a beta thalessemia
WO2015143046A2 (en) 2014-03-18 2015-09-24 Sangamo Biosciences, Inc. Methods and compositions for regulation of zinc finger protein expression
EP3929279A1 (en) 2014-03-18 2021-12-29 Sangamo Therapeutics, Inc. Methods and compositions for regulation of zinc finger protein expression
WO2015168108A2 (en) 2014-04-28 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treating cancer using nucleic targeting mdm2 or mycn
WO2015168605A1 (en) 2014-05-01 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treatment of disorders in the front of the eye utilizing nucleic acid molecules
WO2015171932A1 (en) 2014-05-08 2015-11-12 Sangamo Biosciences, Inc. Methods and compositions for treating huntington's disease
US9487802B2 (en) 2014-05-30 2016-11-08 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods to treat latent viral infections
WO2015184268A1 (en) 2014-05-30 2015-12-03 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods of delivering treatments for latent viral infections
US10066241B2 (en) 2014-05-30 2018-09-04 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods of delivering treatments for latent viral infections
EP3674408A1 (en) 2014-06-16 2020-07-01 The Johns Hopkins University Compositions and methods for the expression of crispr guide rnas
US10195280B2 (en) 2014-07-15 2019-02-05 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US10792362B2 (en) 2014-07-15 2020-10-06 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US10738278B2 (en) 2014-07-15 2020-08-11 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
US11872285B2 (en) 2014-07-15 2024-01-16 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US11254922B2 (en) 2014-07-28 2022-02-22 Sk Innovation Co., Ltd. Isoprene synthase and method of preparing isoprene using thereof
US10287565B2 (en) 2014-07-28 2019-05-14 Sk Innovation Co., Ltd. Isoprene synthase and method of preparing isoprene using thereof
US11578343B2 (en) 2014-07-30 2023-02-14 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
EP3686279A1 (en) 2014-08-17 2020-07-29 The Broad Institute, Inc. Genome editing using cas9 nickases
US11926828B2 (en) 2014-09-05 2024-03-12 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting TYR or MMP1
WO2016037071A2 (en) 2014-09-05 2016-03-10 Rxi Pharmaceuticals Corporation Methods for treating aging and skin disorders using nucleic acids targeting tyr or mmp1
US10900039B2 (en) 2014-09-05 2021-01-26 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting Tyr or MMP1
WO2016044416A1 (en) 2014-09-16 2016-03-24 Sangamo Biosciences, Inc. Methods and compositions for nuclease-mediated genome engineering and correction in hematopoietic stem cells
EP3878948A1 (en) 2014-09-16 2021-09-15 Sangamo Therapeutics, Inc. Methods and compositions for nuclease-mediated genome engineering and correction in hematopoietic stem cells
US11459557B2 (en) 2014-09-24 2022-10-04 The Broad Institute, Inc. Use and production of CHD8+/− transgenic animals with behavioral phenotypes characteristic of autism spectrum disorder
WO2016049163A2 (en) 2014-09-24 2016-03-31 The Broad Institute Inc. Use and production of chd8+/- transgenic animals with behavioral phenotypes characteristic of autism spectrum disorder
US11197467B2 (en) 2014-09-24 2021-12-14 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-cas systems and compositions for modeling mutations in leukocytes
US11124796B2 (en) 2014-09-24 2021-09-21 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for modeling competition of multiple cancer mutations in vivo
WO2016049251A1 (en) 2014-09-24 2016-03-31 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for modeling mutations in leukocytes
US20170298297A1 (en) * 2014-10-01 2017-10-19 Yeda Research And Development Co. Ltd. Liposomes-containing antifouling compositions and uses thereof
WO2016094867A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Protected guide rnas (pgrnas)
WO2016094872A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Dead guides for crispr transcription factors
WO2016094874A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Escorted and functionalized guides for crispr-cas systems
EP3889260A1 (en) 2014-12-12 2021-10-06 The Broad Institute, Inc. Protected guide rnas (pgrnas)
EP3985115A1 (en) 2014-12-12 2022-04-20 The Broad Institute, Inc. Protected guide rnas (pgrnas)
WO2016106236A1 (en) 2014-12-23 2016-06-30 The Broad Institute Inc. Rna-targeting system
EP3702456A1 (en) 2014-12-24 2020-09-02 The Broad Institute, Inc. Crispr having or associated with destabilization domains
WO2016106244A1 (en) 2014-12-24 2016-06-30 The Broad Institute Inc. Crispr having or associated with destabilization domains
WO2016108926A1 (en) 2014-12-30 2016-07-07 The Broad Institute Inc. Crispr mediated in vivo modeling and genetic screening of tumor growth and metastasis
EP4257690A2 (en) 2015-01-28 2023-10-11 Caribou Biosciences, Inc. Crispr hybrid dna/rna polynucleotides and methods of use
WO2016123230A1 (en) 2015-01-28 2016-08-04 Pioneer Hi-Bred International, Inc. Crispr hybrid dna/rna polynucleotides and methods of use
EP4335918A2 (en) 2015-04-03 2024-03-13 Dana-Farber Cancer Institute, Inc. Composition and methods of genome editing of b-cells
WO2017176806A1 (en) 2015-04-03 2017-10-12 Dana-Farber Cancer Institute, Inc. Composition and methods of genome editing of b cells
WO2016161446A1 (en) 2015-04-03 2016-10-06 Dana-Farber Cancer Institute, Inc. Composition and methods of genome editing of b-cells
WO2016186772A2 (en) 2015-05-16 2016-11-24 Genzyme Corporation Gene editing of deep intronic mutations
WO2016196388A1 (en) 2015-05-29 2016-12-08 Juno Therapeutics, Inc. Composition and methods for regulating inhibitory interactions in genetically engineered cells
US10117911B2 (en) 2015-05-29 2018-11-06 Agenovir Corporation Compositions and methods to treat herpes simplex virus infections
US11643669B2 (en) 2015-06-17 2023-05-09 Massachusetts Institute Of Technology CRISPR mediated recording of cellular events
WO2016205728A1 (en) 2015-06-17 2016-12-22 Massachusetts Institute Of Technology Crispr mediated recording of cellular events
EP4159856A1 (en) 2015-06-18 2023-04-05 The Broad Institute, Inc. Novel crispr enzymes and systems
EP3666895A1 (en) 2015-06-18 2020-06-17 The Broad Institute, Inc. Novel crispr enzymes and systems
WO2016205749A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Novel crispr enzymes and systems
US11180751B2 (en) 2015-06-18 2021-11-23 The Broad Institute, Inc. CRISPR enzymes and systems
WO2016205711A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Novel crispr enzymes and systems
WO2016205764A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Novel crispr enzymes and systems
EP3009511A2 (en) 2015-06-18 2016-04-20 The Broad Institute, Inc. Novel crispr enzymes and systems
EP3502253A1 (en) 2015-06-18 2019-06-26 The Broad Institute Inc. Novel crispr enzymes and systems
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
EP3862005A1 (en) 2015-07-06 2021-08-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (sod1)
US10808247B2 (en) 2015-07-06 2020-10-20 Phio Pharmaceuticals Corp. Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
WO2017011519A1 (en) 2015-07-13 2017-01-19 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
US10786533B2 (en) 2015-07-15 2020-09-29 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
WO2017009489A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Topical compositions
WO2017009485A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Topical compositions
WO2017009486A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Topical compositions
WO2017009488A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Topical compositions
EP3747425A1 (en) 2015-07-16 2020-12-09 Nuritas Limited Anti-inflammatory peptides, and uses thereof
WO2017009491A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Peptides for use in promoting transport of glucose
EP3932418A2 (en) 2015-07-16 2022-01-05 Nuritas Limited Peptides for use in promoting transport of glucose
WO2017009487A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Topical compositions
WO2017009492A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Antibacterial peptides, and uses thereof
EP4219521A2 (en) 2015-07-16 2023-08-02 Nuritas Limited Cellular growth promoting peptides derived from rice proteins (oryza sativa) and uses thereof
EP3954700A1 (en) 2015-07-16 2022-02-16 Nuritas Limited Anti-inflammatory peptides derived from rice proteins (oryza sativa) and uses thereof
EP3998276A2 (en) 2015-07-16 2022-05-18 Nuritas Limited Growth promoting peptides and uses thereof
EP3590523A1 (en) 2015-07-16 2020-01-08 Nuritas Limited Growth promoting peptides and uses thereof
WO2017009490A1 (en) 2015-07-16 2017-01-19 Nuritas Limited Growth promoting peptides and uses thereof
US11559486B2 (en) 2015-07-22 2023-01-24 Acrotech Biopharma, LLC Ready-to-use formulation for Vincristine Sulfate Liposome Injection
WO2017024317A2 (en) 2015-08-06 2017-02-09 Dana-Farber Cancer Institute, Inc. Methods to induce targeted protein degradation through bifunctional molecules
US11692182B2 (en) 2015-10-09 2023-07-04 Monsanto Technology Llc RNA-guided DNA nucleases and uses thereof
US11021707B2 (en) 2015-10-19 2021-06-01 Phio Pharmaceuticals Corp. Reduced size self-delivering nucleic acid compounds targeting long non-coding RNA
WO2017070151A1 (en) 2015-10-19 2017-04-27 Rxi Pharmaceuticals Corporation Reduced size self-delivering nucleic acid compounds targeting long non-coding rna
WO2017070605A1 (en) 2015-10-22 2017-04-27 The Broad Institute Inc. Type vi-b crispr enzymes and systems
US11214780B2 (en) 2015-10-23 2022-01-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2017106657A1 (en) 2015-12-18 2017-06-22 The Broad Institute Inc. Novel crispr enzymes and systems
WO2017106537A2 (en) 2015-12-18 2017-06-22 Sangamo Biosciences, Inc. Targeted disruption of the mhc cell receptor
WO2017106528A2 (en) 2015-12-18 2017-06-22 Sangamo Biosciences, Inc. Targeted disruption of the t cell receptor
WO2017123757A1 (en) 2016-01-15 2017-07-20 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of neurologic disease
US10724020B2 (en) 2016-02-02 2020-07-28 Sangamo Therapeutics, Inc. Compositions for linking DNA-binding domains and cleavage domains
EP3769775A2 (en) 2016-02-02 2021-01-27 Sangamo Therapeutics, Inc. Compositions for linking dna-binding domains and cleavage domains
US11920169B2 (en) 2016-02-02 2024-03-05 Sangamo Therapeutics, Inc. Compositions for linking DNA-binding domains and cleavage domains
WO2017165788A1 (en) 2016-03-24 2017-09-28 Marquette University Quantitative flagellar fluorescent markers and standards
WO2017189308A1 (en) 2016-04-19 2017-11-02 The Broad Institute Inc. Novel crispr enzymes and systems
WO2017184768A1 (en) 2016-04-19 2017-10-26 The Broad Institute Inc. Novel crispr enzymes and systems
WO2017184786A1 (en) 2016-04-19 2017-10-26 The Broad Institute Inc. Cpf1 complexes with reduced indel activity
WO2018229521A1 (en) 2016-06-16 2018-12-20 Oslo Universitetssykehus Hf Improved gene editing
US11788083B2 (en) 2016-06-17 2023-10-17 The Broad Institute, Inc. Type VI CRISPR orthologs and systems
WO2017219027A1 (en) 2016-06-17 2017-12-21 The Broad Institute Inc. Type vi crispr orthologs and systems
WO2018005873A1 (en) 2016-06-29 2018-01-04 The Broad Institute Inc. Crispr-cas systems having destabilization domain
US11766488B2 (en) 2016-07-05 2023-09-26 The Johns Hopkins University Compositions and methods comprising improvements of CRISPR guide RNAS using the H1 promoter
WO2018013840A1 (en) 2016-07-13 2018-01-18 Vertex Pharmaceuticals Incorporated Methods, compositions and kits for increasing genome editing efficiency
EP4219462A1 (en) 2016-07-13 2023-08-02 Vertex Pharmaceuticals Incorporated Methods, compositions and kits for increasing genome editing efficiency
US11674158B2 (en) 2016-07-15 2023-06-13 Salk Institute For Biological Studies Methods and compositions for genome editing in non-dividing cells
EP4321623A2 (en) 2016-07-15 2024-02-14 Salk Institute for Biological Studies Methods and compositions for genome editing in non-dividing cells
WO2018013932A1 (en) 2016-07-15 2018-01-18 Salk Institute For Biological Studies Methods and compositions for genome editing in non-dividing cells
WO2018014936A1 (en) 2016-07-18 2018-01-25 Nuritas Limited Topical compositions
US11702651B2 (en) 2016-08-03 2023-07-18 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
WO2018029586A1 (en) 2016-08-07 2018-02-15 Novartis Ag Mrna-mediated immunization methods
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
WO2018035387A1 (en) 2016-08-17 2018-02-22 The Broad Institute, Inc. Novel crispr enzymes and systems
WO2018035388A1 (en) 2016-08-17 2018-02-22 The Broad Institute, Inc. Novel crispr enzymes and systems
WO2018039440A1 (en) 2016-08-24 2018-03-01 Sangamo Therapeutics, Inc. Regulation of gene expression using engineered nucleases
EP3964573A1 (en) 2016-08-24 2022-03-09 Sangamo Therapeutics, Inc. Engineered target specific nucleases
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
WO2018039448A1 (en) 2016-08-24 2018-03-01 Sangamo Therapeutics, Inc. Engineered target specific nucleases
EP3995574A1 (en) 2016-08-24 2022-05-11 Sangamo Therapeutics, Inc. Regulation of gene expression using engineered nucleases
WO2018067826A1 (en) 2016-10-05 2018-04-12 Cellular Dynamics International, Inc. Generating mature lineages from induced pluripotent stem cells with mecp2 disruption
EP4190335A1 (en) 2016-10-13 2023-06-07 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
US11896615B2 (en) 2016-10-13 2024-02-13 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
WO2018071873A2 (en) 2016-10-13 2018-04-19 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
US11219695B2 (en) 2016-10-20 2022-01-11 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of Fabry disease
WO2018081775A1 (en) 2016-10-31 2018-05-03 Sangamo Therapeutics, Inc. Gene correction of scid-related genes in hematopoietic stem and progenitor cells
US10011848B2 (en) 2016-11-09 2018-07-03 City University Of Hong Kong System and method for delivery of substance into mammalian cells
WO2018102612A1 (en) 2016-12-02 2018-06-07 Juno Therapeutics, Inc. Engineered b cells and related compositions and methods
US11793833B2 (en) 2016-12-02 2023-10-24 Juno Therapeutics, Inc. Engineered B cells and related compositions and methods
EP4129321A1 (en) 2016-12-05 2023-02-08 Nuritas Limited Compositions comprising peptide wkdeagkplvk
EP3329905A1 (en) 2016-12-05 2018-06-06 Nuritas Limited Topical cosmetic compositions comprising an oligopeptide against anti-aging of the skin
WO2018104346A1 (en) 2016-12-05 2018-06-14 Nuritas Limited Compositions comprising peptide wkdeagkplvk
EP3329930A1 (en) 2016-12-05 2018-06-06 Nuritas Limited Pharmaceuctical compositions
EP4276187A2 (en) 2016-12-08 2023-11-15 Case Western Reserve University Methods and compositions for enhancing functional myelin production
WO2018106782A1 (en) 2016-12-08 2018-06-14 Case Western Reserve University Methods and compositions for enhancing functional myelin production
WO2018119060A1 (en) 2016-12-20 2018-06-28 Bristol-Myers Squibb Company Methods for increasing the efficiency of homology directed repair (hdr) in the cellular genome
EP4286523A2 (en) 2016-12-20 2023-12-06 Bristol-Myers Squibb Company Methods for increasing the efficiency of homology directed repair (hdr) in the cellular genome
US11820969B2 (en) 2016-12-23 2023-11-21 President And Fellows Of Harvard College Editing of CCR2 receptor gene to protect against HIV infection
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
WO2018170333A1 (en) 2017-03-15 2018-09-20 The Broad Institute, Inc. Novel cas13b orthologues crispr enzymes and systems
US11739308B2 (en) 2017-03-15 2023-08-29 The Broad Institute, Inc. Cas13b orthologues CRISPR enzymes and systems
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
WO2018187688A1 (en) 2017-04-07 2018-10-11 Massachusetts Institute Of Technology Methods to spatially profile protease activity in tissue and sections
WO2018191388A1 (en) 2017-04-12 2018-10-18 The Broad Institute, Inc. Novel type vi crispr orthologs and systems
US11872262B2 (en) 2017-05-09 2024-01-16 Vib Vzw Means and methods for treating bacterial infections
WO2018208910A1 (en) 2017-05-09 2018-11-15 The Broad Institute Inc. Gut microbiome function predicts response to anti-integrin biologic therapy in inflammatory bowel diseases
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
WO2018234370A1 (en) 2017-06-20 2018-12-27 Institut Curie Immune cells defective for suv39h1
EP3828264A1 (en) 2017-06-20 2021-06-02 Institut Curie Immune cells defective for suv39h1
EP3916086A1 (en) 2017-06-23 2021-12-01 Inscripta, Inc. Nucleic acid-guided nucleases
WO2019005884A1 (en) 2017-06-26 2019-01-03 The Broad Institute, Inc. Crispr/cas-adenine deaminase based compositions, systems, and methods for targeted nucleic acid editing
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
WO2019023680A1 (en) 2017-07-28 2019-01-31 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (pace)
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11932884B2 (en) 2017-08-30 2024-03-19 President And Fellows Of Harvard College High efficiency base editors comprising Gam
WO2019060746A1 (en) 2017-09-21 2019-03-28 The Broad Institute, Inc. Systems, methods, and compositions for targeted nucleic acid editing
WO2019075292A1 (en) 2017-10-12 2019-04-18 Massachusetts Institute Of Technology Prostate cancer protease nanosensors and uses thereof
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
US11578334B2 (en) 2017-10-25 2023-02-14 Monsanto Technology Llc Targeted endonuclease activity of the RNA-guided endonuclease CasX in eukaryotes
US11851679B2 (en) 2017-11-01 2023-12-26 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019089982A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019100023A1 (en) 2017-11-17 2019-05-23 Iovance Biotherapeutics, Inc. Til expansion from fine needle aspirates and small biopsies
WO2019103857A1 (en) 2017-11-22 2019-05-31 Iovance Biotherapeutics, Inc. Expansion of peripheral blood lymphocytes (pbls) from peripheral blood
US11713446B2 (en) 2018-01-08 2023-08-01 Iovance Biotherapeutics, Inc. Processes for generating TIL products enriched for tumor antigen-specific T-cells
WO2019136456A1 (en) 2018-01-08 2019-07-11 Iovance Biotherapeutics, Inc. Processes for generating til products enriched for tumor antigen-specific t-cells
WO2019136459A1 (en) 2018-01-08 2019-07-11 Iovance Biotherapeutics, Inc. Processes for generating til products enriched for tumor antigen-specific t-cells
WO2019143677A1 (en) 2018-01-17 2019-07-25 Vertex Pharmaceuticals Incorporated Quinoxalinone compounds, compositions, methods, and kits for increasing genome editing efficiency
WO2019143675A1 (en) 2018-01-17 2019-07-25 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
WO2019143678A1 (en) 2018-01-17 2019-07-25 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
WO2019160383A1 (en) 2018-02-19 2019-08-22 고려대학교 산학협력단 Vaccine comprising epitope of heat shock protein, and use thereof
WO2019173332A1 (en) 2018-03-05 2019-09-12 Massachusetts Institute Of Technology Inhalable nanosensors with volatile reporters and uses thereof
US10945955B2 (en) 2018-04-25 2021-03-16 University Of Massachusetts Artificial exosome composition and related methods
WO2019226953A1 (en) 2018-05-23 2019-11-28 The Broad Institute, Inc. Base editors and uses thereof
WO2019236566A1 (en) 2018-06-05 2019-12-12 Lifeedit, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2020033601A1 (en) 2018-08-07 2020-02-13 The Broad Institute, Inc. Novel cas12b enzymes and systems
WO2020041380A1 (en) 2018-08-20 2020-02-27 The Broad Institute, Inc. Methods and compositions for optochemical control of crispr-cas9
EP4234570A2 (en) 2018-09-18 2023-08-30 Sangamo Therapeutics, Inc. Programmed cell death 1 (pd1) specific nucleases
WO2020068920A2 (en) 2018-09-25 2020-04-02 Massachusetts Institute Of Technology Lung protease nanosensors and uses thereof
WO2020069029A1 (en) 2018-09-26 2020-04-02 Emendobio Inc. Novel crispr nucleases
WO2020069373A1 (en) 2018-09-28 2020-04-02 President And Fellows Of Harvard College Cellular reprogramming to reverse aging and promote organ and tissue regeneration
WO2020065062A1 (en) 2018-09-28 2020-04-02 Wageningen Universiteit Off-target activity inhibitors for guided endonucleases
WO2020079033A1 (en) 2018-10-15 2020-04-23 Fondazione Telethon Genome editing methods and constructs
WO2020081635A2 (en) 2018-10-16 2020-04-23 Massachusetts Institute Of Technology Renal clearable nanocatalysts for disease monitoring
WO2020092453A1 (en) 2018-10-29 2020-05-07 The Broad Institute, Inc. Nucleobase editors comprising geocas9 and uses thereof
WO2020096986A2 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Selection of improved tumor reactive t-cells
WO2020096927A1 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Expansion of tils utilizing akt pathway inhibitors
WO2020096989A1 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients refractory for anti-pd-1 antibody
WO2020096988A2 (en) 2018-11-05 2020-05-14 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
WO2020102659A1 (en) 2018-11-15 2020-05-22 The Broad Institute, Inc. G-to-t base editors and uses thereof
WO2020131862A1 (en) 2018-12-17 2020-06-25 The Broad Institute, Inc. Crispr-associated transposase systems and methods of use thereof
WO2020131547A1 (en) 2018-12-19 2020-06-25 Iovance Biotherapeutics, Inc. Methods of expanding tumor infiltrating lymphocytes using engineered cytokine receptor pairs and uses thereof
EP4339286A2 (en) 2018-12-27 2024-03-20 LifeEDIT Therapeutics, Inc. Polypeptides useful for gene editing and methods of use
WO2020139783A2 (en) 2018-12-27 2020-07-02 Lifeedit, Inc. Polypeptides useful for gene editing and methods of use
WO2020150560A1 (en) 2019-01-17 2020-07-23 Massachusetts Institute Of Technology Sensors for detecting and imaging of cancer metastasis
WO2020163379A1 (en) 2019-02-05 2020-08-13 Emendobio Inc. Crispr compositions and methods for promoting gene editing of ribosomal protein s19 (rps19) gene
US11866469B2 (en) 2019-02-06 2024-01-09 Klogenix Llc DNA binding proteins and uses thereof
WO2020163307A1 (en) 2019-02-06 2020-08-13 Emendobio Inc. New engineered high fidelity cas9
WO2020172343A2 (en) 2019-02-19 2020-08-27 Massachusetts Institute Of Technology Methods for treating injuries
WO2020180733A1 (en) 2019-03-01 2020-09-10 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes from liquid tumors and therapeutic uses thereof
WO2020181178A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. T:a to a:t base editing through thymine alkylation
WO2020181195A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. T:a to a:t base editing through adenine excision
WO2020181193A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. T:a to a:t base editing through adenosine methylation
WO2020181180A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. A:t to c:g base editors and uses thereof
WO2020181202A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. A:t to t:a base editing through adenine deamination and oxidation
WO2020183197A1 (en) 2019-03-14 2020-09-17 Ucl Business Ltd Minimal promoter
WO2020191102A1 (en) 2019-03-18 2020-09-24 The Broad Institute, Inc. Type vii crispr proteins and systems
DE112020001342T5 (en) 2019-03-19 2022-01-13 President and Fellows of Harvard College Methods and compositions for editing nucleotide sequences
DE112020001339T5 (en) 2019-03-19 2022-01-13 President and Fellows of Harvard College METHOD AND COMPOSITION FOR EDITING NUCLEOTIDE SEQUENCES
WO2020191171A1 (en) 2019-03-19 2020-09-24 The Broad Institute, Inc. Methods and compositions for editing nucleotide sequences
US11643652B2 (en) 2019-03-19 2023-05-09 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
DE112020001306T5 (en) 2019-03-19 2022-01-27 Massachusetts Institute Of Technology METHODS AND COMPOSITIONS FOR EDITING NUCLEOTIDE SEQUENCES
WO2020191153A2 (en) 2019-03-19 2020-09-24 The Broad Institute, Inc. Methods and compositions for editing nucleotide sequences
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11795452B2 (en) 2019-03-19 2023-10-24 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
WO2020205838A1 (en) 2019-04-02 2020-10-08 Sangamo Therapeutics, Inc. Methods for the treatment of beta-thalassemia
EP4335925A2 (en) 2019-04-09 2024-03-13 The Regents of the University of California Long-lasting analgesia via targeted in vivo epigenetic repression
WO2020210751A1 (en) 2019-04-12 2020-10-15 The Broad Institute, Inc. System for genome editing
WO2020214842A1 (en) 2019-04-17 2020-10-22 The Broad Institute, Inc. Adenine base editors with reduced off-target effects
WO2020223539A1 (en) 2019-04-30 2020-11-05 The Broad Institute, Inc. Methods and compositions for barcoding nucleic acid libraries and cell populations
WO2020232029A1 (en) 2019-05-13 2020-11-19 Iovance Biotherapeutics, Inc. Methods and compositions for selecting tumor infiltrating lymphocytes and uses of the same in immunotherapy
WO2020232271A1 (en) 2019-05-14 2020-11-19 The Broad Institute, Inc. Compositions and methods for targeting multinucleated cells
WO2020236972A2 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Non-class i multi-component nucleic acid targeting systems
WO2020236982A1 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Aav delivery of nucleobase editors
WO2020243371A1 (en) 2019-05-28 2020-12-03 Massachusetts Institute Of Technology Methods and compositions for modulating immune responses
US11447790B2 (en) 2019-05-29 2022-09-20 Altria Client Services Llc Compositions and methods for producing tobacco plants and products having reduced or eliminated suckers
WO2020243485A1 (en) 2019-05-29 2020-12-03 Massachusetts Institute Of Technology Hiv-1 specific immunogen compositions and methods of use
WO2020243276A1 (en) 2019-05-29 2020-12-03 Altria Client Services Llc Compositions and methods for producing tobacco plants and products having reduced or eliminated suckers
EP4166671A1 (en) 2019-05-29 2023-04-19 Altria Client Services LLC Compositions and methods for producing tobacco plants and products having reduced or eliminated suckers
WO2021001534A1 (en) 2019-07-03 2021-01-07 Wageningen Universiteit Crispr type v-u1 system from mycobacterium mucogenicum and uses thereof
WO2021013950A1 (en) 2019-07-23 2021-01-28 Mnemo Therapeutics Immune cells defective for suv39h1
WO2021025750A1 (en) 2019-08-08 2021-02-11 The Broad Institute, Inc. Base editors with diversified targeting scope
WO2021030344A1 (en) 2019-08-12 2021-02-18 Lifeedit, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2021030666A1 (en) 2019-08-15 2021-02-18 The Broad Institute, Inc. Base editing by transglycosylation
WO2021034717A1 (en) 2019-08-16 2021-02-25 Massachusetts Institute Of Technology Targeted trans-splicing using crispr/cas13
WO2021045292A1 (en) 2019-09-03 2021-03-11 주식회사 제노포커스 Crm197 protein expression method
US11827893B2 (en) 2019-10-10 2023-11-28 Altria Client Services Llc Pale yellow locus and its applications in tobacco
WO2021072328A1 (en) 2019-10-10 2021-04-15 The Broad Institute, Inc. Methods and compositions for prime editing rna
WO2021072248A2 (en) 2019-10-10 2021-04-15 Altria Client Services Llc Pale yellow locus and its applications in tobacco
WO2021087361A1 (en) 2019-11-01 2021-05-06 Sangamo Therapeutics, Inc. Zinc finger nuclease variants for treating or preventing lysosomal storage diseases
WO2021087366A1 (en) 2019-11-01 2021-05-06 Sangamo Therapeutics, Inc. Compositions and methods for genome engineering
WO2021092464A2 (en) 2019-11-08 2021-05-14 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides targeting bromodomain containing protein 4 (brd4) for immunotherapy
WO2021108717A2 (en) 2019-11-26 2021-06-03 The Broad Institute, Inc Systems and methods for evaluating cas9-independent off-target editing of nucleic acids
WO2021118990A1 (en) 2019-12-11 2021-06-17 Iovance Biotherapeutics, Inc. Processes for the production of tumor infiltrating lymphocytes (tils) and methods of using the same
WO2021126435A1 (en) 2019-12-20 2021-06-24 Novarock Biotherapeutics, Ltd. Anti-interleukin-23 p19 antibodies and methods of use thereof
WO2021138247A1 (en) 2019-12-30 2021-07-08 LifeEDIT Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2021138537A1 (en) 2019-12-31 2021-07-08 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides with improved systemic delivery
WO2021148788A1 (en) 2020-01-22 2021-07-29 Ucl Business Ltd Engineered immune cells
GB202000934D0 (en) 2020-01-22 2020-03-04 Ucl Business Ltd Engineered immune cells
WO2021155065A1 (en) 2020-01-28 2021-08-05 The Broad Institute, Inc. Base editors, compositions, and methods for modifying the mitochondrial genome
WO2021158999A1 (en) 2020-02-05 2021-08-12 The Broad Institute, Inc. Gene editing methods for treating spinal muscular atrophy
WO2021158921A2 (en) 2020-02-05 2021-08-12 The Broad Institute, Inc. Adenine base editors and uses thereof
WO2021163191A1 (en) 2020-02-10 2021-08-19 Board Of Regents, The University Of Texas System Methods for rapid cloning and expression of hla class i cells
WO2021167687A1 (en) 2020-02-18 2021-08-26 Massachusetts Institute Of Technology Multiplexed in vivo disease sensing with nucleic acid-barcoded reporters
WO2021217002A1 (en) 2020-04-24 2021-10-28 Lifeedit Therapeutics, Inc . Rna-guided nucleases and active fragments and variants thereof and methods of use
EP4242306A2 (en) 2020-04-24 2023-09-13 Lifeedit Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2021222318A1 (en) 2020-04-28 2021-11-04 The Broad Institute, Inc. Targeted base editing of the ush2a gene
WO2021226061A1 (en) 2020-05-04 2021-11-11 Iovance Biotherapeutics, Inc. Processes for production of tumor infiltrating lymphocytes and uses of the same in immunotherapy
WO2021224416A1 (en) 2020-05-06 2021-11-11 Cellectis S.A. Methods to genetically modify cells for delivery of therapeutic proteins
WO2021224395A1 (en) 2020-05-06 2021-11-11 Cellectis S.A. Methods for targeted insertion of exogenous sequences in cellular genomes
DE112021002672T5 (en) 2020-05-08 2023-04-13 President And Fellows Of Harvard College METHODS AND COMPOSITIONS FOR EDIT BOTH STRANDS SIMULTANEOUSLY OF A DOUBLE STRANDED NUCLEOTIDE TARGET SEQUENCE
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
WO2021231437A1 (en) 2020-05-11 2021-11-18 LifeEDIT Therapeutics, Inc. Rna-guided nucleic acid binding proteins and active fragments and variants thereof and methods of use
EP3939596A1 (en) 2020-07-14 2022-01-19 Bimeda Animal Health Limited A composition for treating helminth infestation in a mammal
WO2022013323A1 (en) 2020-07-14 2022-01-20 Bimeda Animal Health Limited A composition for treating helminth infestation in a non-human mammal
WO2022015969A1 (en) 2020-07-15 2022-01-20 LifeEDIT Therapeutics, Inc. Uracil stabilizing proteins and active fragments and variants thereof and methods of use
WO2022023576A1 (en) 2020-07-30 2022-02-03 Institut Curie Immune cells defective for socs1
WO2022034374A2 (en) 2020-08-11 2022-02-17 University Of Oslo Improved gene editing
WO2022046760A2 (en) 2020-08-25 2022-03-03 Kite Pharma, Inc. T cells with improved functionality
WO2022051591A2 (en) 2020-09-04 2022-03-10 Novarock Biotherapeutics, Ltd. Nectin-4 antibodies and uses thereof
WO2022056254A2 (en) 2020-09-11 2022-03-17 LifeEDIT Therapeutics, Inc. Dna modifying enzymes and active fragments and variants thereof and methods of use
WO2022060927A1 (en) 2020-09-17 2022-03-24 Northwestern University Engineered mammalian genetic circuits and methods of using the same
WO2022081015A1 (en) 2020-10-16 2022-04-21 Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis Anti-tumor immunity induces the presentation of aberrant peptides
WO2022150790A2 (en) 2021-01-11 2022-07-14 The Broad Institute, Inc. Prime editor variants, constructs, and methods for enhancing prime editing efficiency and precision
WO2022164319A1 (en) 2021-01-29 2022-08-04 Wageningen Universiteit Crispr-associated base-editing of the complementary strand
WO2022169839A1 (en) 2021-02-03 2022-08-11 Altria Client Services Llc Increasing trichome density and improving transport of metabolites in plant trichomes
WO2022169837A1 (en) 2021-02-03 2022-08-11 Altria Client Services Llc Tissue-specific promoters in plants
WO2022169836A1 (en) 2021-02-03 2022-08-11 Altria Client Services Llc Terpene production in plants
WO2022192789A1 (en) 2021-03-12 2022-09-15 Synthego Corporation Genetically modified cells expressing antigen-containing fusion proteins and uses thereof
WO2022204093A1 (en) 2021-03-22 2022-09-29 LifeEDIT Therapeutics, Inc. Dna modifyng enzymes and active fragments and variants thereof and methods of use
WO2022219008A1 (en) 2021-04-14 2022-10-20 University College Cork - National University Of Ireland, Cork Treatment of cerebrovascular events and neurological disorders
WO2022219168A1 (en) 2021-04-14 2022-10-20 University College Cork - National University Of Ireland, Cork Psg1 for use in the treatment of osteoarthritis
WO2022243565A1 (en) 2021-05-21 2022-11-24 Cellectis S.A. Enhancing efficacy of t-cell-mediated immunotherapy by modulating cancer-associated fibroblasts in solid tumors
WO2022261394A1 (en) 2021-06-11 2022-12-15 LifeEDIT Therapeutics, Inc. Rna polymerase iii promoters and methods of use
WO2022261509A1 (en) 2021-06-11 2022-12-15 The Broad Institute, Inc. Improved cytosine to guanine base editors
WO2023015265A2 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides
WO2023015264A1 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Immunotherapy of cancer utilizing natural killer cells treated with chemically modified oligonucleotides
WO2023015309A2 (en) 2021-08-06 2023-02-09 The Broad Institute, Inc. Improved prime editors and methods of use
WO2023055239A1 (en) 2021-10-01 2023-04-06 Koninklijke Nederlandse Akademie Van Wetenschappen Mammalian cardiac regeneration
WO2023076898A1 (en) 2021-10-25 2023-05-04 The Broad Institute, Inc. Methods and compositions for editing a genome with prime editing and a recombinase
WO2023077148A1 (en) 2021-11-01 2023-05-04 Tome Biosciences, Inc. Single construct platform for simultaneous delivery of gene editing machinery and nucleic acid cargo
US11649444B1 (en) 2021-11-02 2023-05-16 Huidagene Therapeutics Co., Ltd. CRISPR-CAS12i systems
WO2023080788A1 (en) 2021-11-05 2023-05-11 Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis Tryptophan depletion induces production and presentation of tryptophan to phenylalanine substitutions
WO2023096494A1 (en) 2021-11-26 2023-06-01 Synvolux Ip B.V. Conserved coronavirus t cell epitopes
WO2023122764A1 (en) 2021-12-22 2023-06-29 Tome Biosciences, Inc. Co-delivery of a gene editor construct and a donor template
WO2023126458A1 (en) 2021-12-28 2023-07-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
WO2023139557A1 (en) 2022-01-24 2023-07-27 LifeEDIT Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2023150637A1 (en) 2022-02-02 2023-08-10 Inscripta, Inc. Nucleic acid-guided nickase fusion proteins
WO2023196802A1 (en) 2022-04-04 2023-10-12 The Broad Institute, Inc. Cas9 variants having non-canonical pam specificities and uses thereof
WO2023196818A1 (en) 2022-04-04 2023-10-12 The Regents Of The University Of California Genetic complementation compositions and methods
WO2023196851A1 (en) 2022-04-06 2023-10-12 President And Fellows Of Harvard College Reversing aging of the central nervous system
WO2023205744A1 (en) 2022-04-20 2023-10-26 Tome Biosciences, Inc. Programmable gene insertion compositions
WO2023212715A1 (en) 2022-04-28 2023-11-02 The Broad Institute, Inc. Aav vectors encoding base editors and uses thereof
WO2023215831A1 (en) 2022-05-04 2023-11-09 Tome Biosciences, Inc. Guide rna compositions for programmable gene insertion
WO2023220392A1 (en) 2022-05-13 2023-11-16 Northwestern University Synthetic modular extracellular sensors that employ natural receptor ligand-binding domains
WO2023220457A1 (en) 2022-05-13 2023-11-16 Northwestern University Receptor engagement-mediated enhancement of biologics delivery
WO2023220459A1 (en) 2022-05-13 2023-11-16 Northwestern University Active loading of cargo entity into lipid bilayer particles using dimerization domains
WO2023222928A2 (en) 2022-05-20 2023-11-23 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023225670A2 (en) 2022-05-20 2023-11-23 Tome Biosciences, Inc. Ex vivo programmable gene insertion
WO2023230433A1 (en) 2022-05-23 2023-11-30 Altria Client Services Llc Methods and compositions for regulating alkaloids in tobacco field
WO2023230613A1 (en) 2022-05-27 2023-11-30 The Broad Institute, Inc. Improved mitochondrial base editors and methods for editing mitochondrial dna
WO2023240137A1 (en) 2022-06-08 2023-12-14 The Board Institute, Inc. Evolved cas14a1 variants, compositions, and methods of making and using same in genome editing
WO2024003334A1 (en) 2022-06-30 2024-01-04 Cellectis S.A. Enhancing safety of t-cell-mediated immunotherapy
WO2024015383A1 (en) 2022-07-12 2024-01-18 Northwestern University Engineered hypoxia biosensors and methods of using the same
WO2024020587A2 (en) 2022-07-22 2024-01-25 Tome Biosciences, Inc. Pleiopluripotent stem cell programmable gene insertion
WO2024031038A1 (en) 2022-08-05 2024-02-08 Altria Client Services Llc Methods and compositions for regulating alkaloids in tobacco
WO2024033901A1 (en) 2022-08-12 2024-02-15 LifeEDIT Therapeutics, Inc. Rna-guided nucleases and active fragments and variants thereof and methods of use
WO2024040083A1 (en) 2022-08-16 2024-02-22 The Broad Institute, Inc. Evolved cytosine deaminases and methods of editing dna using same
WO2024042489A1 (en) 2022-08-25 2024-02-29 LifeEDIT Therapeutics, Inc. Chemical modification of guide rnas with locked nucleic acid for rna guided nuclease-mediated gene editing
WO2024047587A1 (en) 2022-08-31 2024-03-07 Regel Therapeutics, Inc. Cas-phi compositions and methods of use
US11959094B2 (en) 2023-05-02 2024-04-16 Salk Institute For Biological Studies Methods and compositions for genome editing in non-dividing cells

Also Published As

Publication number Publication date
AU7979491A (en) 1991-11-27

Similar Documents

Publication Publication Date Title
WO1991017424A1 (en) Intracellular delivery of biologically active substances by means of self-assembling lipid complexes
CA2079814C (en) Cationic lipids for intracellular delivery of biologically active molecules
US5753613A (en) Compositions for the introduction of polyanionic materials into cells
US5981501A (en) Methods for encapsulating plasmids in lipid bilayers
CA2263705C (en) Novel sandwich liposome complexes comprising a biologically active agent
US7288266B2 (en) Liposome complexes for increased systemic delivery
US6110745A (en) Preparation of lipid-nucleic acid particles using a solvent extraction and direct hydration method
US5785992A (en) Compositions for the introduction of polyanionic materials into cells
US8242089B2 (en) Sphingolipids polyalkylamine conjugates for use in transfection
US6770291B2 (en) Liposome complexes for increased systemic delivery
US20030099697A1 (en) Amphoteric liposomes and their use
WO1998007408A9 (en) Novel liposome complexes for increased systemic delivery
US7001614B2 (en) Liposome complexes for increased systemic delivery
KOSHIZAKA et al. Novel liposomes for efficient transfection of β-galactosidase gene into COS-1 cells
US6156338A (en) Method for preparing compositions for transferring nucleic acids
KR100373845B1 (en) Cationic lipids and method for preparing the same
RU2002112224A (en) Cationic Virosome DOSPER
Zarif et al. Lipid-Based Vehicles for Gene Delivery-Concept, Achievements and Future Development

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LU NL SE

NENP Non-entry into the national phase

Ref country code: CA